1
|
Zhang C, Yin X, Jiang J, Wang P, Wang Y. Downregulation of GFPT2 enhances cisplatin chemotherapy sensitivity in STK11/KRAS mutant non-small cell lung cancer by regulating the hexosamine biosynthesis pathway, resisting tumor growth. Cytokine 2025; 191:156943. [PMID: 40253947 DOI: 10.1016/j.cyto.2025.156943] [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: 01/03/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE To explore the role of GFPT2 in the sensitivity of STK11/KRAS lung cancer cells to cisplatin chemotherapy, and its underlying mechanism. MATERIALS & METHODS A549 and H460 cells were used to analyze the effect of GFPT2 on cisplatin chemotherapy sensitivity, as both carry KRAS mutations and H460 has LKB1 inactivation mutations, meeting the requirements of a KRAS/LKB1 co mutant tumor model. The levels of UDP-GlcNAc, OGT, OGA, and O-GlcNAc in the HBP pathway were also determined. To verify the potential role of HBP, we added OGT inhibitors. In vivo, we constructed a nude mouse model bearing A549 tumor to further validate the results of in vitro cell experiments. RESULTS GFPT2 silencing can significantly inhibit cell proliferation, invasion, and migration, promote cell apoptosis, and enhance the effect of cisplatin (p < 0.05). After OSMI-1 processing, GFPT2 enhances O-GlcNAc modification levels via the OGT-mediated HBP, thereby decreasing the sensitivity of STK11/KRAS mutant cells to cisplatin chemotherapy. In addition, GFPT2 silencing enhances the chemotherapy sensitivity of cisplatin and inhibits tumor growth, while overexpression of GFPT2 weakens this effect (p < 0.05). The above results provide new targets and combination therapy options for the clinical treatment of KRAS/LKB1 mutant lung cancer. CONCLUSION Our study found that inhibiting GFPT2 can enhance the chemotherapy sensitivity of cisplatin to STK11/KRAS/LKB1 mutant NSCLCs cells through the OGT mediated HBP pathway, filling a key gap in the chemotherapy resistance mechanism of KRAS/LKB1 mutant lung cancer.
Collapse
Affiliation(s)
- Cheng Zhang
- Thoracic Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Xuelei Yin
- Yantai Keyou Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Jun Jiang
- Key Laboratory of Genetics Research and Evaluation of the National Drug Administration, Shandong Food and Drug Inspection and Research Institute, Jinan, Shandong, China
| | - Peng Wang
- Ministry of Scientific and Technological Innovation, Yantai Hi-tech Industrial Development Zone, Yantai, Shandong, China
| | - Yirong Wang
- Department of Radiotherapy, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
2
|
Yang Y, Li Q, Chu LT, Lin X, Chen H, Chen L, Tang J, Zeng T. Autophagy in cholangiocarcinoma: a comprehensive review about roles and regulatory mechanisms. Clin Transl Oncol 2025; 27:2391-2400. [PMID: 39585591 DOI: 10.1007/s12094-024-03797-7] [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: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
The role of autophagy in cholangiocarcinogenesis and its development is intricate. Autophagy has a dual role in cholangiocarcinoma, and understanding the function and mechanism of autophagy in cholangiocarcinoma is pivotal in guiding therapeutic approaches to its treatment in clinical settings. Recent studies have revealed that autophagy is involved in the complex biological behavior of cholangiocarcinoma. In this review, we have summarized the genes and drugs that would promote or inhibit autophagy, leading to change in cellular behaviors of cholangiocarcinoma, including apoptosis, proliferation, invasion and migration, and influence its cellular drug resistance. In addition, we concluded the signaling pathways modulating autophagy in cholangiocarcinoma cells, including PI3K/AKT/mTOR,p38MAPK,AMPK/mTOR,LKB1-AMPK, and AKT/WNK1, and ERK signaling pathways, which subsequently impacting apoptosis, death, migration, invasion, and proliferation. In conclusion, we would like that we can provide ideas for future cholangiocarcinoma treatment by comprehensively summarizing the latest studies on the relationship between autophagy and cholangiocarcinoma, including the factors affecting autophagy and related signaling pathways.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Qiuyan Li
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Lok Ting Chu
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Xiaocong Lin
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Helian Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Linsong Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Jinjing Tang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China.
| |
Collapse
|
3
|
Yao J, Song S, Liu T, Wang J, Li C, Liu J, Yuan Y, Zhao H. Isoguanosine-Induced ER Stress via AMPK Enhances Chemosensitivity in OSCC. J Dent Res 2025; 104:668-678. [PMID: 40071313 DOI: 10.1177/00220345241303168] [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: 03/17/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck; however, the efficacy of existing treatment is limited and new effective strategies need to be explored. Our previous work demonstrates that isoguanosine (isoG) is a promising nucleoside molecule with superior self-assembly capability and significant anti-OSCC potential. However, the antitumor mechanism of isoG remains unclear. In this study, we reveal that the antiproliferative effect of isoG is mediated by its cellular metabolite, isoguanosine 5'-monophosphate (isoGMP), which induces excessive endoplasmic reticulum (ER) stress and cell death through adenosine monophosphate-activated protein kinase (AMPK) activation. IsoG activates AMPK and induces ER stress at low concentrations, with minimal impact on cell viability at these concentrations. To further explore the therapeutic potential of isoG, we investigated its role in modulating chemosensitivity. Our findings show that AMPK activation enhances the sensitivity of OSCC cells to 5-fluorouracil (5-FU), and the combination of isoG and 5-FU exhibits a synergistic anticancer effect. Building on the self-assembly characteristics of isoG, we developed an innovative treatment platform by introducing dynamic borate ester bonds to form an isoguanosine-phenylenediboronic acid-isoguanosine (isoGPBisoG) structure. When combined with 5-FU, this platform achieved remarkable therapeutic efficacy in 2 OSCC cell-derived xenograft models, with tumor inhibition rates of 71.0% and 56.6%, respectively, compared with control. These findings establish isoG as a potent enhancer of chemotherapeutic efficacy in OSCC via AMPK activation. More importantly, the isoGPBisoG and 5-FU combination represents a significant paradigm of a synergistic therapy platform. This novel approach offers a promising direction for the development of more effective OSCC treatments.
Collapse
Affiliation(s)
- J Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - T Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - C Li
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu, People's Republic of China
| | - J Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Shteinfer-Kuzmine A, Moyal MM, Karunanithi Nivedita A, Trishna S, Nadir A, Tripathi S, Shoshan-Barmatz V. Metformin-Induced Apoptosis Is Mediated Through Mitochondrial VDAC1. Pharmaceuticals (Basel) 2025; 18:757. [PMID: 40430574 DOI: 10.3390/ph18050757] [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: 03/04/2025] [Revised: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Besides diabetes mellitus, metformin has been identified as a potential therapeutic agent for treating various other conditions that include various cancers, cardiovascular diseases, neurodegenerative diseases, and aging. In cancer, metformin increased apoptotic cell death, while inhibiting it in neurodegenerative diseases. Thus, different modes of metformin action at the molecular level have been proposed. Methods: In this study, we present the mitochondria and the VDAC1 (voltage-dependent anion channel) as a potential target of metformin. Results: Metformin induces VDAC1 overexpression, its oligomerization, and subsequent apoptosis. Metformin analogs phenformin and buformin at much lower concentrations also induce VDAC1 overexpression, oligomerization, and cell death. We demonstrate the interaction of metformin with purified VDAC1, which inhibited its channel conduction in a voltage-dependent manner. Metformin bound to the synthetic VDAC1-N-terminal peptide and binding to this domain was also found by its molecular docking with VDAC1. Moreover, we demonstrated metformin binding to purified hexokinases (HK-I) with a 400-fold lower metformin concentration than that required for cell death induction. In cells, metformin induced HK-I detachment from the mitochondrial VDAC1. Lastly, metformin increased the expression of NLRP3 and ASC and induced their co-localization, suggesting inflammasome activation. Conclusions: The results suggest that VDAC1 is a target for metformin and its analogs, and this is associated with metformin's adverse effects on many diseases.
Collapse
Affiliation(s)
- Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Meital M Moyal
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | - Sweta Trishna
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Almog Nadir
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Shubhandra Tripathi
- Department of Chemistry, Indian Institute of Technology, Kanpur 208016, India
| | - Varda Shoshan-Barmatz
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
5
|
Tao ZH, Han JX, Xu J, Zhao E, Wang M, Wang Z, Lin XL, Xiao XY, Hong J, Chen H, Chen YX, Chen HM, Fang JY. Screening of patient-derived organoids identifies mitophagy as a cell-intrinsic vulnerability in colorectal cancer during statin treatment. Cell Rep Med 2025; 6:102039. [PMID: 40154491 PMCID: PMC12047522 DOI: 10.1016/j.xcrm.2025.102039] [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: 09/22/2024] [Revised: 01/26/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Statins, commonly used to lower cholesterol, are associated with improved prognosis in colorectal cancer (CRC), though their effectiveness varies. This study investigates the anti-cancer effects of atorvastatin in CRC using patient-derived organoids (PDOs) and PDO-derived xenograft (PDOX) models. Our findings reveal that atorvastatin induces mitochondrial dysfunction, leading to apoptosis in cancer cells. In response, cancer cells induce mitophagy to clear damaged mitochondria, enhancing survival and reducing statin efficacy. Analysis of a clinical cohort confirms mitophagy's role in diminishing statin effectiveness. Importantly, inhibiting mitophagy significantly enhances the anti-cancer effects of atorvastatin in CRC PDOs, xenograft models, and azoxymethane (AOM)-dextran sulfate sodium (DSS) mouse models. These findings identify mitophagy as a critical pro-survival mechanism in CRC during statin treatment, providing insights into the variable responses observed in epidemiological studies. Targeting this vulnerability through combination therapy can elicit potent therapeutic responses.
Collapse
Affiliation(s)
- Zhi-Hang Tao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ji-Xuan Han
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Enhao Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Lin Lin
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiu-Ying Xiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Min Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Lorenz NI, Sauer B, Urban H, Weinem JB, Parmar BS, Zeiner PS, Strecker MI, Schulte D, Mittelbronn M, Alekseeva T, Sevenich L, Harter PN, Münch C, Steinbach JP, Luger AL, Heiland DH, Ronellenfitsch MW. AMP-activated protein kinase mediates adaptation of glioblastoma cells to conditions of the tumor microenvironment. J Exp Clin Cancer Res 2025; 44:104. [PMID: 40122814 PMCID: PMC11931870 DOI: 10.1186/s13046-025-03346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/22/2025] [Indexed: 03/25/2025] Open
Abstract
AMP-activated protein kinase (AMPK) is an energy sensor that regulates cellular metabolic activity. We hypothesized that in glioblastoma (GB), AMPK plays a pivotal role in balancing metabolism under conditions of the tumor microenvironment with fluctuating and often low nutrient and oxygen availability. Impairment of this network could thus interfere with tumor progression. AMPK activity was modulated genetically by CRISPR/Cas9-based double knockout (DKO) of the catalytic α1 and α2 subunits in human GB cells and effects were confirmed by pharmacological AMPK inhibition using BAY3827 and an inactive control compound in primary GB cell cultures. We found that metabolic adaptation of GB cells under energy stress conditions (hypoxia, glucose deprivation) was dependent on AMPK and accordingly that AMPK DKO cells were more vulnerable to glucose deprivation or inhibition of glycolysis and sensitized to hypoxia-induced cell death. This effect was rescued by reexpression of the AMPK α2 subunit. Similar results were observed using the selective pharmacological AMPK inhibitor BAY3827. Mitochondrial biogenesis was regulated AMPK-dependently with a reduced mitochondrial mass and mitochondrial membrane potential in AMPK DKO GB cells. In vivo, AMPK DKO GB cells showed impaired tumor growth and tumor formation in CAM assays as well as in an orthotopic glioma mouse model. Our study highlights the importance of AMPK for GB cell adaptation towards energy depletion and emphasizes the role of AMPK for tumor formation in vivo. Moreover, we identified mitochondria as central downstream effectors of AMPK signaling. The development of AMPK inhibitors could open opportunities for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Nadja I Lorenz
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Benedikt Sauer
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
| | - Hans Urban
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jan-Béla Weinem
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Bhavesh S Parmar
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
| | - Pia S Zeiner
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Maja I Strecker
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Luxembourg Centre of Neuropathology (LCNP), Dudelange, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur- Alzette, Luxembourg
- Department of Life Science and Medicine (DLSM), University of Luxembourg, Esch-sur- Alzette, Luxembourg
| | - Tijna Alekseeva
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Lisa Sevenich
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Patrick N Harter
- Center for Neuropathology and Prion Research, Faculty of Medicine, Ludwig-Maximilians- University of Munich, Munich, Germany
| | - Christian Münch
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- Institute of Molecular Systems Medicine, Goethe University, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Frankfurt am Main, Germany
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Anna-Luisa Luger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, Freiburg, Germany
- Department of Neurosurgery, University Clinic, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael W Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
7
|
Middleton G, Robbins HL, Fletcher P, Savage J, Mehmi M, Summers Y, Greystoke A, Steele N, Popat S, Jain P, Spicer J, Cave J, Shaw P, Gilligan D, Power D, Fennell D, Bajracharya M, McBride DJ, Maheswari U, Frankell AM, Swanton C, Beggs AD, Billingham L. A phase II trial of mTORC1/2 inhibition in STK11 deficient non small cell lung cancer. NPJ Precis Oncol 2025; 9:67. [PMID: 40069402 PMCID: PMC11897347 DOI: 10.1038/s41698-025-00838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
There are no current stratified medicine options for STK11-deficient NSCLC. STK11 loss mediates mTORC activation, GLUT1 up-regulation and increased glycolysis. This metabolic reprogramming might represent a therapeutic vulnerability targetable with mTORC1/2 inhibition. In arm B2 of the National Lung Matrix Trial 54 patients with NSCLC received vistusertib, of which 49 were STK11-deficient (30 with KRAS mutation (B2D), 19 without (B2S)). Objective response (OR) and durable clinical benefit (DCB) rates with 95% credible intervals (CrI) were estimated from posterior probability distributions generated using Bayesian beta-binomial conjugate analysis. In B2D, 2 per-protocol patients obtained OR (estimated true OR rate (95%CrI) 9.8% (2.4-24.3). Estimates of true DCB rate (95%CrI): B2D 24.4% (11.1-42.3), B2S 14.6% (3.6-34.7). Overall, vistusertib cannot be recommended in this context. Longitudinal ctDNA analysis demonstrates enrichment of SMARCA4 mutations post-treatment. In vitro studies show adaptive resistance to mTORC1/2 inhibition via AKT reactivation. (NCT02664935, ISRCTN38344105, EudraCT 2014-000814-73, 10 June 2015).
Collapse
Affiliation(s)
- Gary Middleton
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Helen L Robbins
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Peter Fletcher
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Joshua Savage
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Manita Mehmi
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | | | | | | | | | - Pooja Jain
- St James's University Hospital, Leeds, UK
| | - James Spicer
- King's College London, Guy's Hospital, London, UK
| | - Judith Cave
- Southampton University Hospitals NHS Trust, Southampton, UK
| | | | | | | | | | | | | | | | - Alexander M Frankell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Andrew D Beggs
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Cancer & Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| |
Collapse
|
8
|
Sasaki N, Homme M, Murayama T, Osaki T, Tenma T, An T, Takegami Y, Tani T, Gedeon PC, Kobayashi Y, Cañadas I, Barbie DA, Yao R, Kitajima S. RNA sensing induced by chromosome missegregation augments anti-tumor immunity. Mol Cell 2025; 85:770-786.e7. [PMID: 39706184 PMCID: PMC11888943 DOI: 10.1016/j.molcel.2024.11.025] [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: 03/18/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Viral mimicry driven by endogenous double-stranded RNA (dsRNA) stimulates innate and adaptive immune responses. However, the mechanisms that regulate dsRNA-forming transcripts during cancer therapy remain unclear. Here, we demonstrate that dsRNA is significantly accumulated in cancer cells following pharmacologic induction of micronuclei, stimulating mitochondrial antiviral signaling (MAVS)-mediated dsRNA sensing in conjunction with the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway. Activation of cytosolic dsRNA sensing cooperates with double-stranded DNA (dsDNA) sensing to upregulate immune cell migration and antigen-presenting machinery. Tracing of dsRNA-sequences reveals that dsRNA-forming transcripts are predominantly generated from non-exonic regions, particularly in locations proximal to genes exhibiting high chromatin accessibility. Activation of this pathway by pulsed monopolar spindle 1 (MPS1) inhibitor treatment, which potently induces micronuclei formation, upregulates cytoplasmic dsRNA sensing and thus promotes anti-tumor immunity mediated by cytotoxic lymphocyte activation in vivo. Collectively, our findings uncover a mechanism in which dsRNA sensing cooperates with dsDNA sensing to boost immune responses, offering an approach to enhance the efficacy of cancer therapies targeting genomic instability.
Collapse
MESH Headings
- Humans
- RNA, Double-Stranded/genetics
- RNA, Double-Stranded/immunology
- RNA, Double-Stranded/metabolism
- Animals
- Nucleotidyltransferases/genetics
- Nucleotidyltransferases/metabolism
- Nucleotidyltransferases/immunology
- Cell Line, Tumor
- Mice
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/immunology
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/pathology
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/antagonists & inhibitors
- Mice, Inbred C57BL
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/immunology
- Signal Transduction
- Micronuclei, Chromosome-Defective
- Immunity, Innate
- Lymphocyte Activation
- Genomic Instability
Collapse
Affiliation(s)
- Nobunari Sasaki
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Mizuki Homme
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Takahiko Murayama
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Tatsuya Osaki
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo 153-8505, Japan
| | - Toshiyuki Tenma
- Respiratory Center, Asahikawa Medical University Hospital, Asahikawa 078-8510, Japan
| | - Tadaichi An
- DNAFORM Precision Gene Technologies, Yokohama, Kanagawa 230-0051, Japan
| | - Yujiro Takegami
- DNAFORM Precision Gene Technologies, Yokohama, Kanagawa 230-0051, Japan
| | - Tetsuo Tani
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Patrick C Gedeon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yoshihisa Kobayashi
- Division of Molecular Pathology, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Shunsuke Kitajima
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
9
|
Zhong ZT, Wang XY, Pan Y, Zhou K, Chen JH, Gao YQ, Dai B, Zhou ZL, Wang RQ. AMPK: An energy sensor for non-small cell lung cancer progression and treatment. Pharmacol Res 2025; 212:107592. [PMID: 39805353 DOI: 10.1016/j.phrs.2025.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Lung cancer (LC) is the leading cause of cancer-related morbidity and mortality in China, with non-small cell lung cancer (NSCLC) accounting for 85 % of the overall lung cancer cases. AMP-activated protein kinase (AMPK) is a key regulator of energy balance and homeostasis, and its dysregulation is a common feature in various malignancies, particularly in NSCLC with mutations in Liver kinase B1 (LKB1). Studies have shown that the AMPK signalling pathway has a dual role in NSCLC progression, both inhibiting and promoting the progression of malignant tumours. Therefore, drugs targeting the AMPK signalling pathway may hold significant promise for therapeutic application in NSCLC. This review aims to examine the manifestations and mechanisms by which AMPK and its associated signalling molecules influence NSCLC progression and treatment. Firstly, we discuss the critical importance of AMPK within the mutational context of NSCLC. Secondly, we summarise the drugs and related substances that modulate the AMPK signalling pathway in NSCLC and evaluate the evidence from preclinical studies on combination AMPK-targeted therapies to address the issue of drug resistance in NSCLC under current clinical treatments. In summary, this paper highlights the critical importance of developing AMPK-targeted drugs to enhance therapeutic efficacy in NSCLC, as well as the potential for applying these drugs in clinical therapy to overcome drug resistance.
Collapse
Affiliation(s)
- Zhi-Ting Zhong
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China; College of Pharmacy, Jinan University, Guangzhou, China
| | - Xu-Yan Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ying Pan
- Department of Oncology, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ke Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Jing-Hui Chen
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Yu-Qi Gao
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan City, Guangdong Province 528200, China.
| | - Zhi-Ling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Rui-Qi Wang
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| |
Collapse
|
10
|
Tan I, Chothani S, Lim HH, Lam KP. Alu-Sc-mediated exonization generated a mitochondrial LKB1 gene variant found only in higher order primates. Sci Rep 2025; 15:3360. [PMID: 39870744 PMCID: PMC11772596 DOI: 10.1038/s41598-025-86789-z] [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: 06/27/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
The tumor suppressor LKB1/STK11 plays important roles in regulating cellular metabolism and stress responses and its mutations are associated with various cancers. We recently identified a novel exon 1b within intron 1 of human LKB1/STK11, which generates an alternatively spliced, mitochondria-targeting LKB1 isoform important for regulating mitochondrial oxidative stress. Here we examined the formation of this novel exon 1b and uncovered its relatively late emergence during evolution. Analyses of putative exon 1b genomic sequences within the primate superfamily indicated that the exonization of LKB1/STK11 exon 1b was mediated by the conserved retrotransposable element Alu-Sc. While putative exon 1b sequences are recognizable in most members of the primate family from New World Monkeys onwards, characteristically functional LKB1/STK11 exon 1b, with translation start and 5' and 3' splice sites, could only be found in greater apes and human, and interestingly, correlates with their increased body mass and longevity development.
Collapse
Affiliation(s)
- Ivan Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore, 138648, Singapore
| | | | - Hong-Hwa Lim
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore, 138648, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore, 138648, Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
11
|
Ma L, Zhang J, Dai Z, Liao P, Guan J, Luo Z. Top 100 most-cited articles on apoptosis of non-small cell lung cancer over the past two decades: a bibliometrics analysis. Front Immunol 2025; 15:1512349. [PMID: 39872524 PMCID: PMC11770037 DOI: 10.3389/fimmu.2024.1512349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
Background Recently there has been an increasing number of studies have explored apoptosis mechanisms in lung cancer (LC). However, no researchers have conducted a bibliometric analysis of the most cited articles in this field. Objective To examine the top 100 most influential and cited publications on apoptosis in non-small cell lung cancer (NSCLC) from 2004 to 2023, summarizing research trends and key focus areas. Methods This study utilized the Web of Science Core Database (WOSCC) to research NSCLC apoptosis from 2004 to 2023, using keyword selection and manual screening for article searches. Bibliometrix package of R software 4.3.1 was used to generate distribution statistics for the top ten institutions, journals and authors. Citespace6.2. R6 was used to create the visualization maps for keyword co-occurrence and clustering. VOSviewer1.6.19 was used to conduct cluster analysis of publishing countries (regions), with data exported to SCImago Graphica for geographic visualization and cooperation analysis. VOSviewer1.6.19 was used to produced co-citation maps of institutions, journals, authors, and references. Results From 2004 to 2023, 13316 articles were retrieved, and the top 100 most cited were chosen. These were authored by 934 individuals from 269 institutions across 18 countries and appeared in 45 journals. Citations ranged from 150 to 1,389, with a median of 209.5. The most influential articles appeared in 2005 and 2007 (n=13). The leading countries (regions), institutions, journals and authors were identified as the United States (n=60), Harvard University (n=64), CANCER RESEARCH (n=15), SUN M and YANG JS (n=6). The top five keywords were "expression", "activation", "apoptosis", "pathway" and "gefitinib". This study indicates that enhancing apoptosis through circular RNA regulation and targeting the Nrf2 signaling pathway could become a key research focus in recent years. Conclusion Apoptosis has been the subject of extensive research over many years, particularly in relation to its role in the pathogenesis, diagnosis, and treatment of NSCLC. This study aims to identify highly influential articles and forecast emerging research trends, thereby offering insights into novel therapeutic targets and strategies to overcome drug resistance. The findings are intended to serve as a valuable reference for scholars engaged in this field of study.
Collapse
Affiliation(s)
- Leshi Ma
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Zhang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi Dai
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei Liao
- Department of Oncology, Chongqing Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Chongqing, China
| | - Jieshan Guan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, Shenshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Shanwei, China
| | - Zhijie Luo
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
12
|
Schneider JL, Han S, Nabel CS. Fuel for thought: targeting metabolism in lung cancer. Transl Lung Cancer Res 2024; 13:3692-3717. [PMID: 39830762 PMCID: PMC11736591 DOI: 10.21037/tlcr-24-662] [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] [Received: 07/30/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
For over a century, we have appreciated that the biochemical processes through which micro- and macronutrients are anabolized and catabolized-collectively referred to as "cellular metabolism"-are reprogrammed in malignancies. Cancer cells in lung tumors rewire pathways of nutrient acquisition and metabolism to meet the bioenergetic demands for unchecked proliferation. Advances in precision medicine have ushered in routine genotyping of patient lung tumors, enabling a deeper understanding of the contribution of altered metabolism to tumor biology and patient outcomes. This paradigm shift in thoracic oncology has spawned a new enthusiasm for dissecting oncogenotype-specific metabolic phenotypes and creates opportunity for selective targeting of essential tumor metabolic pathways. In this review, we discuss metabolic states across histologic and molecular subtypes of lung cancers and the additional changes in tumor metabolic pathways that occur during acquired therapeutic resistance. We summarize the clinical investigation of metabolism-specific therapies, addressing successes and limitations to guide the evaluation of these novel strategies in the clinic. Beyond changes in tumor metabolism, we also highlight how non-cellular autonomous processes merit particular consideration when manipulating metabolic processes systemically, such as efforts to disentangle how lung tumor cells influence immunometabolism. As the future of metabolic therapeutics hinges on use of models that faithfully recapitulate metabolic rewiring in lung cancer, we also discuss best practices for harmonizing workflows to capture patient specimens for translational metabolic analyses.
Collapse
Affiliation(s)
- Jaime L. Schneider
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Christopher S. Nabel
- Department of Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
13
|
Lv L, Miao Q, Zhan S, Chen P, Liu W, Lv J, Yan W, Wang D, Liu H, Yin J, Feng J, Song Y, Ye M, Lv T. LKB1 dictates sensitivity to immunotherapy through Skp2-mediated ubiquitination of PD-L1 protein in non-small cell lung cancer. J Immunother Cancer 2024; 12:e009444. [PMID: 39694700 PMCID: PMC11660338 DOI: 10.1136/jitc-2024-009444] [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: 04/15/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Loss-of-function mutations of liver kinase B (LKB1, also termed as STK11 (serine/threonine kinase 11)) are frequently detected in patients with non-small cell lung cancer (NSCLC). The LKB1 mutant NSCLC was refractory to almost all the antitumor treatments, including programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade therapy. Unfortunately, mechanisms underlying resistance to immunotherapy are not fully understood. In this study, we deciphered how LKB1 regulated sensitivity to anti-PD-1/PD-L1 immunotherapy. METHODS We investigated the mutational landscape of LKB1 mutant NSCLC in next generation sequencing (NGS) data sets. Expression of LKB1, PD-L1 and S-phase kinase-associated protein 2 (Skp2) in NSCLC samples were assessed by immunohistochemistry (IHC). The tumor microenvironment (TME) profiling of LKB1 wild type (WT) and mutant NSCLC was performed using fluorescent multiplex IHC. Mass spectrometry and enrichment analysis were used to identify LKB1 interacting proteins. Mechanistic pathways were explored by immunoblotting, ubiquitination assay, cycloheximide chase assay and immunoprecipitation assay. RESULTS By using NGS data sets and histological approaches, we demonstrated that LKB1 status was positively associated with PD-L1 protein expression and conferred a T cell-enriched "hot" TME in NSCLC. Patients with good responses to anti-PD-1/PD-L1 immunotherapy possessed a high level of LKB1 and PD-L1. Skp2 emerged as the molecular hub connecting LKB1 and PD-L1, by which Skp2 catalyzed K63-linked polyubiquitination on K136 and K280 residues to stabilize PD-L1 protein. Inhibition of Skp2 expression by short hairpin RNA or its E3 ligase activity by compound #25 abrogated intact expression of PD-L1 in vitro and generated a T cell-excluded "cold" TME in vivo. Thus, the LKB1-Skp2-PD-L1 regulatory loop was crucial for retaining PD-L1 protein expression and manipulation of this pathway would be a feasible approach for TME remodeling. CONCLUSION LKB1 and Skp2 are required for intact PD-L1 protein expression and TME remodeling in NSCLC. Inhibition of Skp2 resulted in a conversion from "hot" TME to "cold" TME and abrogated therapeutic outcomes of immunotherapy. Screening LKB1 and Skp2 status would be helpful to select recipients who may benefit from anti-PD-1/PD-L1 immunotherapy.
Collapse
MESH Headings
- Humans
- AMP-Activated Protein Kinase Kinases/analysis
- AMP-Activated Protein Kinase Kinases/genetics
- AMP-Activated Protein Kinase Kinases/metabolism
- S-Phase Kinase-Associated Proteins/analysis
- S-Phase Kinase-Associated Proteins/antagonists & inhibitors
- S-Phase Kinase-Associated Proteins/genetics
- S-Phase Kinase-Associated Proteins/metabolism
- B7-H1 Antigen/analysis
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/metabolism
- Ubiquitination
- Loss of Function Mutation
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Immunotherapy/methods
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Specific Pathogen-Free Organisms
- Mice, Inbred C57BL
- Male
- Animals
- Mice
- Xenograft Model Antitumor Assays
- Female
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Ubiquitin-Protein Ligases/analysis
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Cohort Studies
Collapse
Affiliation(s)
- Liting Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qing Miao
- Department of Pharmacy, the 971 Hospital of PLA Navy, Qingdao, China
| | - Sutong Zhan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Peilin Chen
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wei Liu
- Liaoning Kanghui Biotechnology Co. Ltd, Shenyang, China
| | - Jiawen Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wenjie Yan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jie Yin
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jian Feng
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
14
|
Kenny TC, Birsoy K. Mitochondria and Cancer. Cold Spring Harb Perspect Med 2024; 14:a041534. [PMID: 38692736 PMCID: PMC11610758 DOI: 10.1101/cshperspect.a041534] [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: 05/03/2024]
Abstract
Mitochondria are semiautonomous organelles with diverse metabolic and cellular functions including anabolism and energy production through oxidative phosphorylation. Following the pioneering observations of Otto Warburg nearly a century ago, an immense body of work has examined the role of mitochondria in cancer pathogenesis and progression. Here, we summarize the current state of the field, which has coalesced around the position that functional mitochondria are required for cancer cell proliferation. In this review, we discuss how mitochondria influence tumorigenesis by impacting anabolism, intracellular signaling, and the tumor microenvironment. Consistent with their critical functions in tumor formation, mitochondria have become an attractive target for cancer therapy. We provide a comprehensive update on the numerous therapeutic modalities targeting the mitochondria of cancer cells making their way through clinical trials.
Collapse
Affiliation(s)
- Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
15
|
Wessendorf-Rodriguez K, Ruchhoeft ML, Ashley EL, Galvez HM, Murray CW, Huang Y, McGregor GH, Kambhampati S, Shaw RJ, Metallo CM. Modeling compound lipid homeostasis using stable isotope tracing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618599. [PMID: 39463985 PMCID: PMC11507872 DOI: 10.1101/2024.10.16.618599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Lipids represent the most diverse pool of metabolites found in cells, facilitating compartmentation, signaling, and other functions. Dysregulation of lipid metabolism is linked to disease states such as cancer and neurodegeneration. However, limited tools are available for quantifying metabolic fluxes across the lipidome. To directly measure reaction fluxes encompassing compound lipid homeostasis, we applied stable isotope tracing, liquid chromatography-high-resolution mass spectrometry, and network-based isotopologue modeling to non-small cell lung cancer (NSCLC) models. Compound lipid metabolic flux analysis (CL-MFA) enables the concurrent quantitation of fatty acid synthesis, elongation, headgroup assembly, and salvage reactions within virtually any biological system. Here, we resolve liver kinase B1 (LKB1)-mediated regulation of sphingolipid recycling in NSCLC cells and precision-cut lung slice cultures. We also demonstrate that widely used tissue culture conditions drive cells to upregulate fatty acid synthase flux to supraphysiological levels. Finally, we identify previously uncharacterized isozyme specificity of ceramide synthase inhibitors. These results highlight the ability of CL-MFA to quantify lipid cycling in biological systems to discover biological function and elucidate molecular mechanisms in membrane lipid metabolism.
Collapse
|
16
|
Zhong Q, Li D, Yang XP. Progress in antitumor mechanisms and applications of phenformin (Review). Oncol Rep 2024; 52:151. [PMID: 39301645 PMCID: PMC11421015 DOI: 10.3892/or.2024.8810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
Phenformin, a biguanide compound, has attracted increased attention due to its prominent antitumor activity. As a multi‑target agent, the antitumor effects of phenformin involve a wide range of factors, including inhibition of mitochondrial complex I, activation of AMP‑activated protein kinase, impact on the tumor microenvironment, suppression of cancer stem cells and others. In addition, phenformin has been shown to markedly augment the effectiveness of various clinical treatment methods, including radiotherapy, chemotherapy, targeted therapy and immunotherapy. It is noteworthy that breakthrough progress has been made in the treatment of cancer with phenformin with application in clinical trials for the treatment of melanoma. Phenformin not only reduces the lesion area of patients, but also enhances the efficacy of dalafinib/trimetinib. In the present review, the novel breakthroughs in the antitumor effects and mechanisms of phenformin were discussed. In addition, the current review focuses on the clinical development value of phenformin, striving to provide new insights into the future research direction of phenformin in the field of tumor treatment.
Collapse
Affiliation(s)
- Qi Zhong
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Duo Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xiao-Ping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
17
|
Liu W, Jiang Y, Li G, Huang D, Qin T. Oxidative phosphorylation related gene COA6 is a novel indicator for the prognosis and immune response in lung adenocarcinoma. Sci Rep 2024; 14:25970. [PMID: 39472746 PMCID: PMC11522384 DOI: 10.1038/s41598-024-77775-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
Although the initial research focused on glycolysis, mitochondrial oxidative phosphorylation has become a major target of cancer cells. Cytochrome C oxidase assembly factor 6 (COA6) is a conserved assembly factor necessary for complex IV biogenesis. Nevertheless, the clinical predictive value of COA6, especially its correlation with immune cell infiltration in lung adenocarcinoma (LUAD), has not yet been elucidated. COA6 exhibited higher expression levels in LUAD cells and tumor tissues compared to normal tissues. Additionally, heightened COA6 expression was associated with reduced overall survival (OS) and advanced tumor stage. Apart from its role in mitochondrial respiratory processes, COA6 may be involved in the process of antigen binding, immunoglobulin receptor binding. Interestingly, we observed a positive correlation between COA6 expression and tumor mutational burden (TMB), as well as a significant association with decreased immune cell infiltration. COA6 was linked to resistance against gemcitabine and etoposide. We verified that COA6 was highly expressed in LUAD experimentally and cell proliferation was inhibited after COA6 knockdown. Thus, we conclude that the expression of COA6 was correlated with reduced immune cell infiltration. Additionally, COA6 functioned as a biomarker for drug sensitivity and the prognosis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Wenting Liu
- Department of Thoracic Oncology, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yantao Jiang
- Department of Thoracic Oncology, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Guoli Li
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dingzhi Huang
- Department of Thoracic Oncology, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Tingting Qin
- Department of Thoracic Oncology, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
18
|
WANG ZHIGUO, LI KUNLIN, LU CONGHUA, FENG MINGXIA, LIN CAIYU, YIN GUOFANG, LUO DAN, LIU WENYI, JIN KAIYU, DOU YUANYAO, WU DI, ZHENG JIE, ZHANG KEJUN, LI LI, FAN XIANMING. Metformin promotes anti-tumor immunity in STK11 mutant NSCLC through AXIN1-dependent upregulation of multiple nucleotide metabolites. Oncol Res 2024; 32:1637-1648. [PMID: 39308524 PMCID: PMC11413838 DOI: 10.32604/or.2024.052664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/11/2024] [Indexed: 09/25/2024] Open
Abstract
Background Metformin has pleiotropic effects beyond glucose reduction, including tumor inhibition and immune regulation. It enhanced the anti-tumor effects of programmed cell death protein 1 (PD-1) inhibitors in serine/threonine kinase 11 (STK11) mutant non-small cell lung cancer (NSCLC) through an axis inhibition protein 1 (AXIN1)-dependent manner. However, the alterations of tumor metabolism and metabolites upon metformin administration remain unclear. Methods We performed untargeted metabolomics using liquid chromatography (LC)-mass spectrometry (MS)/MS system and conducted cell experiments to verify the results of bioinformatics analysis. Results According to the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database, most metabolites were annotated into metabolism, including nucleotide metabolism. Next, the differentially expressed metabolites in H460 (refers to H460 cells), H460_met (refers to metformin-treated H460 cells), and H460_KO_met (refers to metformin-treated Axin1 -/- H460 cells) were distributed into six clusters based on expression patterns. The clusters with a reversed expression pattern upon metformin treatment were selected for further analysis. We screened out metabolic pathways through KEGG pathway enrichment analysis and found that multiple nucleotide metabolites enriched in this pathway were upregulated. Furthermore, these metabolites enhanced the cytotoxicity of activated T cells on H460 cells in vitro and can activate the stimulator of the interferon genes (STING) pathway independently of AXIN1. Conclusion Relying on AXIN1, metformin upregulated multiple nucleotide metabolites which promoted STING signaling and the killing of activated T cells in STK11 mutant NSCLC, indicating a potential immunotherapeutic strategy for STK11 mutant NSCLC.
Collapse
Affiliation(s)
- ZHIGUO WANG
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - KUNLIN LI
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - CONGHUA LU
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - MINGXIA FENG
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - CAIYU LIN
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - GUOFANG YIN
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - DAN LUO
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - WENYI LIU
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China
| | - KAIYU JIN
- Department of Respiratory Disease, People’s Hospital of Xuyong County, Luzhou, 646000, China
| | - YUANYAO DOU
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - DI WU
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - JIE ZHENG
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - KEJUN ZHANG
- Department of Outpatients, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - LI LI
- Department of Respiratory Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - XIANMING FAN
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
19
|
Krieger MR, Abrahamian M, He KL, Atamdede S, Hakimjavadi H, Momcilovic M, Ostrow D, Maggo SD, Tsang YP, Gai X, Chanfreau GF, Shackelford DB, Teitell MA, Koehler CM. Trafficking of mitochondrial double-stranded RNA from mitochondria to the cytosol. Life Sci Alliance 2024; 7:e202302396. [PMID: 38955468 PMCID: PMC11220484 DOI: 10.26508/lsa.202302396] [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: 09/26/2023] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
In addition to mitochondrial DNA, mitochondrial double-stranded RNA (mtdsRNA) is exported from mitochondria. However, specific channels for RNA transport have not been demonstrated. Here, we begin to characterize channel candidates for mtdsRNA export from the mitochondrial matrix to the cytosol. Down-regulation of SUV3 resulted in the accumulation of mtdsRNAs in the matrix, whereas down-regulation of PNPase resulted in the export of mtdsRNAs to the cytosol. Targeting experiments show that PNPase functions in both the intermembrane space and matrix. Strand-specific sequencing of the double-stranded RNA confirms the mitochondrial origin. Inhibiting or down-regulating outer membrane proteins VDAC1/2 and BAK/BAX or inner membrane proteins PHB1/2 strongly attenuated the export of mtdsRNAs to the cytosol. The cytosolic mtdsRNAs subsequently localized to large granules containing the stress protein TIA-1 and activated the type 1 interferon stress response pathway. Abundant mtdsRNAs were detected in a subset of non-small-cell lung cancer cell lines that were glycolytic, indicating relevance in cancer biology. Thus, we propose that mtdsRNA is a new damage-associated molecular pattern that is exported from mitochondria in a regulated manner.
Collapse
Affiliation(s)
- Matthew R Krieger
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | | | - Kevin L He
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Sean Atamdede
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | | | - Milica Momcilovic
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Dejerianne Ostrow
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Simran Ds Maggo
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yik Pui Tsang
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Xiaowu Gai
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guillaume F Chanfreau
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - David B Shackelford
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Michael A Teitell
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA
- NanoSystems Institute, UCLA, Los Angeles, CA, USA
| | - Carla M Koehler
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
20
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. eLife 2024; 13:RP96992. [PMID: 39213022 PMCID: PMC11364435 DOI: 10.7554/elife.96992] [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] [Indexed: 09/04/2024] Open
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent U.S. Food and Drug Administration approval of covalent inhibitors of KRASG12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRASG12C-driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients who do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRASG12C, efforts are underway to develop effective combination therapies. Here, we report that the inhibition of KRASG12C signaling increases autophagy in KRASG12C-expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRASG12C-driven lung cancer cell proliferation in vitro and superior tumor control in vivo. Additionally, in genetically engineered mouse models of KRASG12C-driven NSCLC, inhibition of either KRASG12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRASG12C in lung cancer.
Collapse
Affiliation(s)
- Phaedra C Ghazi
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Kayla T O'Toole
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Sanjana Srinivas Boggaram
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Michael T Scherzer
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Mark R Silvis
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Yun Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | | | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | - Eric L Snyder
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Pathology, University of UtahSalt Lake CityUnited States
| | - Conan G Kinsey
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Internal Medicine, Division of Medical Oncology, University of UtahSalt Lake CityUnited States
| | - Martin McMahon
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Dermatology, University of UtahSalt Lake CityUnited States
| |
Collapse
|
21
|
Kong Q, Zhu Q, Yang Y, Wang W, Qian J, Chen Y. Current status and trend of mitochondrial research in lung cancer: A bibliometric and visualization analysis. Heliyon 2024; 10:e34442. [PMID: 39144972 PMCID: PMC11320136 DOI: 10.1016/j.heliyon.2024.e34442] [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: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
This study summarizes and analyzes the relationship between mitochondria and the pathogenesis of lung cancer. The related articles in the Web of Science core literature database are searched and collected, and the data are processed by R software, Citespace, VOSviewer, and Excel. A total of 4476 related papers were retrieved, 4476 articles from 20162 co-authors of 3968 institutions in 84 countries and published in 951 journals. Through various bibliometric analysis tools, the relationship between mitochondria and the pathogenesis of lung cancer was analyzed, the previous research results were summarized, and the potential research direction was found.
Collapse
Affiliation(s)
- Qing Kong
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Qingyong Zhu
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Yuxia Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Wei Wang
- Clinical Medical College, Weifang Medical University, Weifang, 261053, PR China
| | - Juan Qian
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Yong Chen
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| |
Collapse
|
22
|
Vernieri C, Ligorio F, Tripathy D, Longo VD. Cyclic fasting-mimicking diet in cancer treatment: Preclinical and clinical evidence. Cell Metab 2024; 36:1644-1667. [PMID: 39059383 DOI: 10.1016/j.cmet.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
In preclinical tumor models, cyclic fasting and fasting-mimicking diets (FMDs) produce antitumor effects that become synergistic when combined with a wide range of standard anticancer treatments while protecting normal tissues from treatment-induced adverse events. More recently, results of phase 1/2 clinical trials showed that cyclic FMD is safe, feasible, and associated with positive metabolic and immunomodulatory effects in patients with different tumor types, thus paving the way for larger clinical trials to investigate FMD anticancer activity in different clinical contexts. Here, we review the tumor-cell-autonomous and immune-system-mediated mechanisms of fasting/FMD antitumor effects, and we critically discuss new metabolic interventions that could synergize with nutrient starvation to boost its anticancer activity and prevent or reverse tumor resistance while minimizing toxicity to patients. Finally, we highlight potential future applications of FMD approaches in combination with standard anticancer strategies as well as strategies to implement the design and conduction of clinical trials.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy.
| | - Francesca Ligorio
- Medical Oncology and Hematology-Oncology Department, University of Milan, 20122 Milan, Italy; Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1354, 1515 Holcombe Blvd, Houston, TX 77030-4009, USA
| | - Valter D Longo
- IFOM ETS, the AIRC Institute of Molecular Oncology, 20139 Milan, Italy; Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
23
|
Ashraf N, Van Nostrand JL. Fine-tuning AMPK in physiology and disease using point-mutant mouse models. Dis Model Mech 2024; 17:dmm050798. [PMID: 39136185 PMCID: PMC11340815 DOI: 10.1242/dmm.050798] [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] [Indexed: 08/17/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase that monitors the cellular energy status to adapt it to the fluctuating nutritional and environmental conditions in an organism. AMPK plays an integral part in a wide array of physiological processes, such as cell growth, autophagy and mitochondrial function, and is implicated in diverse diseases, including cancer, metabolic disorders, cardiovascular diseases and neurodegenerative diseases. AMPK orchestrates many different physiological outcomes by phosphorylating a broad range of downstream substrates. However, the importance of AMPK-mediated regulation of these substrates in vivo remains an ongoing area of investigation to better understand its precise role in cellular and metabolic homeostasis. Here, we provide a comprehensive overview of our understanding of the kinase function of AMPK in vivo, as uncovered from mouse models that harbor phosphorylation mutations in AMPK substrates. We discuss some of the inherent limitations of these mouse models, highlight the broader implications of these studies for understanding human health and disease, and explore the valuable insights gained that could inform future therapeutic strategies for the treatment of metabolic and non-metabolic disorders.
Collapse
Affiliation(s)
- Naghmana Ashraf
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeanine L. Van Nostrand
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
24
|
Gadwal A, Panigrahi P, Khokhar M, Sharma V, Setia P, Vishnoi JR, Elhence P, Purohit P. A critical appraisal of the role of metabolomics in breast cancer research and diagnostics. Clin Chim Acta 2024; 561:119836. [PMID: 38944408 DOI: 10.1016/j.cca.2024.119836] [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: 03/30/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Breast cancer (BC) remains the most prevalent cancer among women worldwide, despite significant advancements in its prevention and treatment. The escalating incidence of BC globally necessitates continued research into novel diagnostic and therapeutic strategies. Metabolomics, a burgeoning field, offers a comprehensive analysis of all metabolites within a cell, tissue, system, or organism, providing crucial insights into the dynamic changes occurring during cancer development and progression. This review focuses on the metabolic alterations associated with BC, highlighting the potential of metabolomics in identifying biomarkers for early detection, diagnosis, treatment and prognosis. Metabolomics studies have revealed distinct metabolic signatures in BC, including alterations in lipid metabolism, amino acid metabolism, and energy metabolism. These metabolic changes not only support the rapid proliferation of cancer cells but also influence the tumour microenvironment and therapeutic response. Furthermore, metabolomics holds great promise in personalized medicine, facilitating the development of tailored treatment strategies based on an individual's metabolic profile. By providing a holistic view of the metabolic changes in BC, metabolomics has the potential to revolutionize our understanding of the disease and improve patient outcomes.
Collapse
Affiliation(s)
- Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Pragyan Panigrahi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Vaishali Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Puneet Setia
- Department of Forensic Medicine and Toxicology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Jeewan Ram Vishnoi
- Department of Oncosurgery, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Poonam Elhence
- Department of Pathology, All India Institute of Medical Sciences, Jodhpur Rajasthan, 342005, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India.
| |
Collapse
|
25
|
Guo W, Lu T, Song Y, Li A, Feng X, Han D, Cao Y, Sun D, Gong X, Li C, Jin R, Du H, Chen K, Xiang J, Hang J, Chen G, Li H. Lymph node metastasis in early invasive lung adenocarcinoma: Prediction model establishment and validation based on genomic profiling and clinicopathologic characteristics. Cancer Med 2024; 13:e70039. [PMID: 39046176 PMCID: PMC11267562 DOI: 10.1002/cam4.70039] [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: 01/06/2024] [Revised: 06/22/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND The presence of lymph node (LN) metastasis directly affects the treatment strategy for lung adenocarcinoma (LUAD). Next-generation sequencing (NGS) has been widely used in patients with advanced LUAD to identify targeted genes, while early detection of pathologic LN metastasis using NGS has not been assessed. METHODS Clinicopathologic features and molecular characteristics of 224 patients from Ruijin Hospital were analyzed to detect factors associated with LN metastases. Another 140 patients from Huashan Hospital were set as a test cohort. RESULTS Twenty-four out of 224 patients were found to have lymph node metastases (10.7%). Pathologic LN-positive tumors showed higher mutant allele tumor heterogeneity (p < 0.05), higher tumor mutation burden (p < 0.001), as well as more frequent KEAP1 (p = 0.001), STK11 (p = 0.004), KRAS (p = 0.007), CTNNB1 (p = 0.017), TP53, and ARID2 mutations (both p = 0.02); whereas low frequency of EGFR mutation (p = 0.005). A predictive nomogram involving male sex, solid tumor morphology, higher T stage, EGFR wild-type, and TP53, STK11, CDKN2A, KEAP1, ARID2, KRAS, SDHA, SPEN, CTNNB1, DICER1 mutations showed outstanding efficiency in both the training cohort (AUC = 0.819) and the test cohort (AUC = 0.780). CONCLUSION This study suggests that the integration of genomic profiling and clinical features identifies early-invasive LUAD patients at higher risk of LN metastasis. Improved identification of LN metastasis is beneficial for the optimization of the patient's therapy decisions.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tong Lu
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yang Song
- Department of Thoracic SurgeryHuashan Hospital, Fudan UniversityShanghaiChina
| | - Anqi Li
- Department of PathologyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xijia Feng
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dingpei Han
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuqin Cao
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Debin Sun
- Genecast Biotechnology Co., LtdWuxiChina
| | | | - Chengqiang Li
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Runsen Jin
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hailei Du
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kai Chen
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Xiang
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junbiao Hang
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Gang Chen
- Department of Thoracic SurgeryHuashan Hospital, Fudan UniversityShanghaiChina
| | - Hecheng Li
- Department of Thoracic SurgeryRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
26
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579200. [PMID: 38370808 PMCID: PMC10871191 DOI: 10.1101/2024.02.06.579200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent FDA approval of covalent inhibitors of KRAS G12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRAS G12C -driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients that do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRAS G12C , efforts are underway to develop effective combination therapies. Here we report that inhibition of KRAS G12C signaling increases autophagy in KRAS G12C expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRAS G12C -driven lung cancer cell proliferation in vitro and superior tumor control in vivo . Additionally, in genetically engineered mouse models of KRAS G12C -driven NSCLC, inhibition of either KRAS G12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRAS G12C in lung cancer.
Collapse
|
27
|
Sun J, Zhang Y, Zhang Q, Hu L, Zhao L, Wang H, Yuan Y, Niu H, Wang D, Zhang H, Liu J, Feng X, Su X, Qiu J, Sun J, Xu H, Zhang C, Wang K, Bi Y, Engleman EG, Shen L. Metabolic regulator LKB1 controls adipose tissue ILC2 PD-1 expression and mitochondrial homeostasis to prevent insulin resistance. Immunity 2024; 57:1289-1305.e9. [PMID: 38772366 DOI: 10.1016/j.immuni.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/06/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Jiping Sun
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youqin Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingbing Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Hu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Linfeng Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Yue Yuan
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Hongshen Niu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dongdi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huasheng Zhang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianyue Liu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xujiao Feng
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaohui Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Catherine Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Kathleen Wang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lei Shen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
28
|
Yao G, Yu S, Hou F, Xiao Z, Li G, Ji X, Wang J. Rab3B enhances the stabilization of DDX6 to promote lung adenocarcinoma aggressiveness. Mol Med 2024; 30:75. [PMID: 38834947 PMCID: PMC11151598 DOI: 10.1186/s10020-024-00848-1] [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: 07/31/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Liver kinase B1 (LKB1) is frequently mutated in lung adenocarcinoma, and its loss contributes to tumor progression. METHODS To identify LKB1 downstream genes that promote lung adenocarcinoma aggressiveness, we performed bioinformatical analysis using publicly available datasets. RESULTS Rab3B was upregulated in LKB1-depleted lung adenocarcinoma cells and suppressed by LKB1 overexpression. CREB protein was enriched at the promoter of Rab3B in lung cancer cells. Silencing of CREB abrogated the upregulation of Rab3B upon LKB1 loss. Immunohistochemistry revealed the elevated expression of Rab3B in lung adenocarcinomas relative to adjacent normal tissues. Upregulation of Rab3B was significantly associated with lymph node metastasis, advanced tumor stage, and reduced overall survival in lung adenocarcinoma patients. Knockdown of Rab3B suppressed and overexpression of Rab3B promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells in vitro. In a mouse xenograft model, Rab3B depletion restrained and Rab3B overexpression augmented the growth of lung adenocarcinoma tumors. Mechanistically, Rab3B interacted with DDX6 and enhanced its protein stability. Ectopic expression of DDX6 significantly promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells. DDX6 knockdown phenocopied the effects of Rab3B depletion on lung adenocarcinoma cells. Additionally, DDX6 overexpression partially rescued the aggressive phenotype of Rab3B-depleted lung adenocarcinoma cells. CONCLUSION LKB1 deficiency promotes Rab3B upregulation via a CREB-dependent manner. Rab3B interacts with and stabilizes DDX6 protein to accelerate lung adenocarcinoma progression. The Rab3B-DDX6 axis may be potential therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Guodong Yao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feng Hou
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Zunyu Xiao
- Department of Imaging, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guangqi Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Xiaobin Ji
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Jigang Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China.
| |
Collapse
|
29
|
Xu K, Lu W, Yu A, Wu H, He J. Effect of the STK11 mutation on therapeutic efficacy and prognosis in patients with non-small cell lung cancer: a comprehensive study based on meta-analyses and bioinformatics analyses. BMC Cancer 2024; 24:491. [PMID: 38632512 PMCID: PMC11025184 DOI: 10.1186/s12885-024-12130-y] [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: 11/23/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND This study aimed to systematically analyze the effect of a serine/threonine kinase (STK11) mutation (STK11mut) on therapeutic efficacy and prognosis in patients with non-small cell lung cancer (NSCLC). METHODS Candidate articles were identified through a search of relevant literature published on or before April 1, 2023, in PubMed, Embase, Cochrane Library, CNKI and Wanfang databases. The extracted and analyzed data included the hazard ratios (HRs) of PFS and OS, the objective response rate (ORR) of immune checkpoint inhibitors (ICIs), and the positive rates of PD-L1 expression. The HR of PFS and OS and the merged ratios were calculated using a meta-analysis. The correlation between STK11mut and clinical characteristics was further analyzed in NSCLC datasets from public databases. RESULTS Fourteen retrospective studies including 4317 patients with NSCLC of whom 605 had STK11mut were included. The meta-analysis revealed that the ORR of ICIs in patients with STK11mut was 10.1% (95%CI 0.9-25.2), and the positive rate of PD-L1 expression was 41.1% (95%CI 25.3-57.0). STK11mut was associated with poor PFS (HR = 1.49, 95%CI 1.28-1.74) and poor OS (HR = 1.44, 95%CI 1.24-1.67). In the bioinformatics analysis, PFS and OS in patients with STK11 alterations were worse than those in patients without alterations (p < 0.001, p = 0.002). Nutlin-3a, 5-fluorouracil, and vinorelbine may have better sensitivity in patients with STK11mut than in those with STK11wt. CONCLUSIONS Patients with STK11-mutant NSCLC had low PD-L1 expression and ORR to ICIs, and their PFS and OS were worse than patients with STK11wt after comprehensive treatment. In the future, more reasonable systematic treatments should be explored for this subgroup of patients with STK11-mutant NSCLC.
Collapse
Affiliation(s)
- Ke Xu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Weinan Lu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Airu Yu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hongwei Wu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jie He
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China.
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Pillai R, LeBoeuf SE, Hao Y, New C, Blum JLE, Rashidfarrokhi A, Huang SM, Bahamon C, Wu WL, Karadal-Ferrena B, Herrera A, Ivanova E, Cross M, Bossowski JP, Ding H, Hayashi M, Rajalingam S, Karakousi T, Sayin VI, Khanna KM, Wong KK, Wild R, Tsirigos A, Poirier JT, Rudin CM, Davidson SM, Koralov SB, Papagiannakopoulos T. Glutamine antagonist DRP-104 suppresses tumor growth and enhances response to checkpoint blockade in KEAP1 mutant lung cancer. SCIENCE ADVANCES 2024; 10:eadm9859. [PMID: 38536921 PMCID: PMC10971495 DOI: 10.1126/sciadv.adm9859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024]
Abstract
Loss-of-function mutations in KEAP1 frequently occur in lung cancer and are associated with poor prognosis and resistance to standard of care treatment, highlighting the need for the development of targeted therapies. We previously showed that KEAP1 mutant tumors consume glutamine to support the metabolic rewiring associated with NRF2-dependent antioxidant production. Here, using preclinical patient-derived xenograft models and antigenic orthotopic lung cancer models, we show that the glutamine antagonist prodrug DRP-104 impairs the growth of KEAP1 mutant tumors. We find that DRP-104 suppresses KEAP1 mutant tumors by inhibiting glutamine-dependent nucleotide synthesis and promoting antitumor T cell responses. Using multimodal single-cell sequencing and ex vivo functional assays, we demonstrate that DRP-104 reverses T cell exhaustion, decreases Tregs, and enhances the function of CD4 and CD8 T cells, culminating in an improved response to anti-PD1 therapy. Our preclinical findings provide compelling evidence that DRP-104, currently in clinical trials, offers a promising therapeutic approach for treating patients with KEAP1 mutant lung cancer.
Collapse
Affiliation(s)
- Ray Pillai
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, NY 10016, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sarah E. LeBoeuf
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, New York University Langone Health, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Connie New
- Departments of Biological Engineering and Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jenna L. E. Blum
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shih Ming Huang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christian Bahamon
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Warren L. Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Burcu Karadal-Ferrena
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alberto Herrera
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ellie Ivanova
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michael Cross
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jozef P. Bossowski
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Hongyu Ding
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Makiko Hayashi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sahith Rajalingam
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Triantafyllia Karakousi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Volkan I. Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41345 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Kamal M. Khanna
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Robert Wild
- Dracen Pharmaceuticals Inc., San Diego, CA 92121, USA
| | - Aristotelis Tsirigos
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - John T. Poirier
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10655, USA
| | - Shawn M. Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
31
|
Skalka GL, Tsakovska M, Murphy DJ. Kinase signalling adaptation supports dysfunctional mitochondria in disease. Front Mol Biosci 2024; 11:1354682. [PMID: 38434478 PMCID: PMC10906720 DOI: 10.3389/fmolb.2024.1354682] [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: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 03/05/2024] Open
Abstract
Mitochondria form a critical control nexus which are essential for maintaining correct tissue homeostasis. An increasing number of studies have identified dysregulation of mitochondria as a driver in cancer. However, which pathways support and promote this adapted mitochondrial function? A key hallmark of cancer is perturbation of kinase signalling pathways. These pathways include mitogen activated protein kinases (MAPK), lipid secondary messenger networks, cyclic-AMP-activated (cAMP)/AMP-activated kinases (AMPK), and Ca2+/calmodulin-dependent protein kinase (CaMK) networks. These signalling pathways have multiple substrates which support initiation and persistence of cancer. Many of these are involved in the regulation of mitochondrial morphology, mitochondrial apoptosis, mitochondrial calcium homeostasis, mitochondrial associated membranes (MAMs), and retrograde ROS signalling. This review will aim to both explore how kinase signalling integrates with these critical mitochondrial pathways and highlight how these systems can be usurped to support the development of disease. In addition, we will identify areas which require further investigation to fully understand the complexities of these regulatory interactions. Overall, this review will emphasize how studying the interaction between kinase signalling and mitochondria improves our understanding of mitochondrial homeostasis and can yield novel therapeutic targets to treat disease.
Collapse
Affiliation(s)
- George L. Skalka
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mina Tsakovska
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Daniel J. Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Scotland Institute, Glasgow, United Kingdom
| |
Collapse
|
32
|
Xiang K, Kunin M, Larafa S, Busch M, Dünker N, Jendrossek V, Matschke J. α-Ketoglutarate supplementation and NAD+ modulation enhance metabolic rewiring and radiosensitization in SLC25A1 inhibited cancer cells. Cell Death Discov 2024; 10:27. [PMID: 38225236 PMCID: PMC10789775 DOI: 10.1038/s41420-024-01805-x] [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: 07/18/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Metabolic rewiring is the result of the increasing demands and proliferation of cancer cells, leading to changes in the biological activities and responses to treatment of cancer cells. The mitochondrial citrate transport protein SLC25A1 is involved in metabolic reprogramming offering a strategy to induce metabolic bottlenecks relevant to radiosensitization through the accumulation of the oncometabolite D-2-hydroxyglutarate (D-2HG) upon SLC25A1 inhibition (SLC25A1i). Previous studies have revealed the comparative effects of SLC25A1i or cell-permeable D-2HG (octyl-D-2HG) treatments on DNA damage induction and repair, as well as on energy metabolism and cellular function, which are crucial for the long-term survival of irradiated cells. Here, α-ketoglutarate (αKG), the precursor of D-2HG, potentiated the effects observed upon SLC25A1i on DNA damage repair, cell function and long-term survival in vitro and in vivo, rendering NCI-H460 cancer cells more vulnerable to ionizing radiation. However, αKG treatment alone had little effect on these phenotypes. In addition, supplementation with nicotinamide (NAM), a precursor of NAD (including NAD+ and NADH), counteracted the effects of SLC25A1i or the combination of SLC25A1i with αKG, highlighting a potential importance of the NAD+/NADH balance on cellular activities relevant to the survival of irradiated cancer cells upon SLC25A1i. Furthermore, inhibition of histone lysine demethylases (KDMs), as a major factor affected upon SLC25A1i, by JIB04 treatment alone or in combination with αKG supplementation phenocopied the broad effects on mitochondrial and cellular function induced by SLC25A1i. Taken together, αKG supplementation potentiated the effects on cellular processes observed upon SLC25A1i and increased the cellular demand for NAD to rebalance the cellular state and ensure survival after irradiation. Future studies will elucidate the underlying metabolic reprogramming induced by SLC25A1i and provide novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Kexu Xiang
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- Department of Gastroenterology, Chongqing University Cancer Hospital, 400030, Chongqing, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, 400030, Chongqing, China
| | - Mikhail Kunin
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Safa Larafa
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Maike Busch
- Center for Translational Neuro- and Behavioral Sciences, Institute of Anatomy II, Department of Neuroanatomy, Medical Faculty, University of Duisburg-Essen, 45147, Essen, Germany
| | - Nicole Dünker
- Center for Translational Neuro- and Behavioral Sciences, Institute of Anatomy II, Department of Neuroanatomy, Medical Faculty, University of Duisburg-Essen, 45147, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- German Cancer Consortium (DKTK) partner site Essen a partnership between DKFZ and University Hospital, Essen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
- German Cancer Consortium (DKTK) partner site Essen a partnership between DKFZ and University Hospital, Essen, Germany.
| |
Collapse
|
33
|
Malawsky DS, Dismuke T, Liu H, Castellino E, Brenman J, Dasgupta B, Tikunov A, Gershon TR. Chronic AMPK inactivation slows SHH medulloblastoma progression by inhibiting mTORC1 signaling and depleting tumor stem cells. iScience 2023; 26:108443. [PMID: 38094249 PMCID: PMC10716552 DOI: 10.1016/j.isci.2023.108443] [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] [Received: 11/21/2022] [Revised: 03/28/2023] [Accepted: 11/08/2023] [Indexed: 01/19/2024] Open
Abstract
We show that inactivating AMPK in a genetic medulloblastoma model depletes tumor stem cells and slows progression. In medulloblastoma, the most common malignant pediatric brain tumor, drug-resistant stem cells co-exist with transit-amplifying cells and terminally differentiated neuronal progeny. Prior studies show that Hk2-dependent glycolysis promotes medulloblastoma progression by suppressing neural differentiation. To determine how the metabolic regulator AMPK affects medulloblastoma growth and differentiation, we inactivated AMPK genetically in medulloblastomas. We bred conditional Prkaa1 and Prkaa2 deletions into medulloblastoma-prone SmoM2 mice and compared SmoM2-driven medulloblastomas with intact or inactivated AMPK. AMPK-inactivation increased event-free survival (EFS) and altered cellular heterogeneity, increasing differentiation and decreasing tumor stem cell populations. Surprisingly, AMPK-inactivation decreased mTORC1 activity and decreased Hk2 expression. Hk2 deletion similarly depleted medulloblastoma stem cells, implicating reduced glycolysis in the AMPK-inactivated phenotype. Our results show that AMPK inactivation disproportionately impairs medulloblastoma stem cell populations typically refractory to conventional therapies.
Collapse
Affiliation(s)
- Daniel Shiloh Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Taylor Dismuke
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Hedi Liu
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Ethan Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jay Brenman
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Biplab Dasgupta
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrey Tikunov
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Center for Neurosciences Research, Children’s Hospital of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Timothy R. Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Center for Neurosciences Research, Children’s Hospital of Atlanta, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
34
|
Zhang L, Zhai BZ, Wu YJ, Wang Y. Recent progress in the development of nanomaterials targeting multiple cancer metabolic pathways: a review of mechanistic approaches for cancer treatment. Drug Deliv 2023; 30:1-18. [PMID: 36597205 PMCID: PMC9943254 DOI: 10.1080/10717544.2022.2144541] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cancer is a very heterogeneous disease, and uncontrolled cell division is the main characteristic of cancer. Cancerous cells need a high nutrition intake to enable aberrant growth and survival. To do so, cancer cells modify metabolic pathways to produce energy and anabolic precursors and preserve redox balance. Due to the importance of metabolic pathways in tumor growth and malignant transformation, metabolic pathways have also been given promising perspectives for cancer treatment, providing more effective treatment strategies, and target-specific with minimum side effects. Metabolism-based therapeutic nanomaterials for targeted cancer treatment are a promising option. Numerous types of nanoparticles (NPs) are employed in the research and analysis of various cancer therapies. The current review focuses on cutting-edge strategies and current cancer therapy methods based on nanomaterials that target various cancer metabolisms. Additionally, it highlighted the primacy of NPs-based cancer therapies over traditional ones, the challenges, and the future potential.
Collapse
Affiliation(s)
- Ling Zhang
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China,CONTACT Ling Zhang Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou310014, Zhejiang, China
| | - Bing-Zhong Zhai
- Hangzhou Municipal Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310021, China
| | - Yue-Jin Wu
- Institute of Food Science and Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China
| | - Yin Wang
- Institute of Food Science and Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China,; Yin Wang Institute of Food Science and Engineering, Hangzhou Medical College, 182 Tianmushan Road, Hangzhou310013, Zhejiang, China
| |
Collapse
|
35
|
Stouras I, Vasileiou M, Kanatas PF, Tziona E, Tsianava C, Theocharis S. Metabolic Profiles of Cancer Stem Cells and Normal Stem Cells and Their Therapeutic Significance. Cells 2023; 12:2686. [PMID: 38067114 PMCID: PMC10705308 DOI: 10.3390/cells12232686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer stem cells (CSCs) are a rare cancer cell population, responsible for the facilitation, progression, and resistance of tumors to therapeutic interventions. This subset of cancer cells with stemness and tumorigenic properties is organized in niches within the tumor microenvironment (TME) and presents altered regulation in a variety of metabolic pathways, including glycolysis, oxidative phosphorylation (OXPHOS), as well as lipid, amino acid, and iron metabolism. CSCs exhibit similarities as well as differences when comparedto normal stem cells, but also possess the ability of metabolic plasticity. In this review, we summarize the metabolic characteristics of normal, non-cancerous stem cells and CSCs. We also highlight the significance and implications of interventions targeting CSC metabolism to potentially achieve more robust clinical responses in the future.
Collapse
Affiliation(s)
- Ioannis Stouras
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Section of Hematology and Medical Oncology, Department of Clinical Therapeutics, General Hospital Alexandra, 11528 Athens, Greece
| | - Maria Vasileiou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Panagiotis F. Kanatas
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Eleni Tziona
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Christina Tsianava
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Rion, Greece;
| | - Stamatis Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| |
Collapse
|
36
|
Shameem M, Jian Bagherpoor A, Nakhi A, Dosa P, Georg G, Kassie F. Mitochondria-targeted metformin (mitomet) inhibits lung cancer in cellular models and in mice by enhancing the generation of reactive oxygen species. Mol Carcinog 2023; 62:1619-1629. [PMID: 37401866 PMCID: PMC10961008 DOI: 10.1002/mc.23603] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/04/2023] [Accepted: 06/08/2023] [Indexed: 07/05/2023]
Abstract
Lung cancer is the leading cause of cancer-related mortality in the United States. Although some epidemiological studies have shown an inverse relationship between the use of metformin, a widely used antidiabetic drug, and the incidence of lung cancer, the real benefits of the drug are unclear as the efficacy is low and the outcomes are quite heterogeneous. To develop a more potent form of metformin, we synthesized mitochondria-targeted metformin (mitomet) and tested its efficacy in in vitro and in vivo models of lung cancer. Mitomet was cytotoxic to transformed bronchial cells and several non-small cell lung cancer (NSCLC) cell lines but relatively safe to normal bronchial cells, and these effects were mediated mainly via induction of mitochondrial reactive oxygen species. Studies using isogenic A549 cells showed that mitomet was selectively toxic to those cells deficient in the tumor suppressor gene LKB1, which is widely mutated in NSCLC. Mitomet also significantly reduced the multiplicity and size of lung tumors induced by a tobacco smoke carcinogen in mice. Overall, our findings showed that mitomet, which was about 1000 and 100 times more potent than metformin, in killing NSCLC cells and reducing the multiplicity and size of lung tumors in mice, respectively, is a promising candidate for the chemoprevention and treatment of lung cancer, in particular against LKB1-deficient lung cancers which are known to be highly aggressive.
Collapse
Affiliation(s)
- Mohammad Shameem
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Ali Nakhi
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Peter Dosa
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Gunda Georg
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Fekadu Kassie
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
37
|
He W, Wang X, Chen M, Li C, Chen W, Pan L, Cui Y, Yu Z, Wu G, Yang Y, Xu M, Dong Z, Ma K, Wang J, He Z. Metformin reduces hepatocarcinogenesis by inducing downregulation of Cyp26a1 and CD8 + T cells. Clin Transl Med 2023; 13:e1465. [PMID: 37997519 PMCID: PMC10668005 DOI: 10.1002/ctm2.1465] [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: 04/14/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly heterogeneous cancer with major challenges in both prevention and therapy. Metformin, adenosine monophosphate-activated protein kinase (AMPK) activator, has been suggested to reduce the incidence of HCC when used for patients with diabetes in preclinical and clinical studies. However, the possible effects of metformin and their mechanisms of action in non-diabetic HCC have not been adequately investigated. METHODS Fah-/- mice were used to construct a liver-injury-induced non-diabetic HCC model for exploring hepatocarcinogenesis and therapeutic potential of metformin. Changes in relevant tumour and biochemical indicators were measured. Bulk and single-cell RNA-sequencing analyses were performed to validate the crucial role of proinflammatory/pro-tumour CD8+ T cells. In vitro and in vivo experiments were performed to confirm Cyp26a1-related antitumour mechanisms of metformin. RESULTS RNA-sequencing analysis showed that chronic liver injury led to significant changes in AMPK-, glucose- and retinol metabolism-related pathways in Fah-/- mice. Metformin prevented the formation of non-diabetic HCC in Fah-/- mice with chronic liver injury. Cyp26a1 ddexpression in hepatocytes was significantly suppressed after metformin treatment. Moreover, downregulation of Cyp26a1 occurred in conjunction with increased levels of all-trans-retinoic acid (atRA), which is involved in the activation of metformin-suppressed hepatocarcinogenesis in Fah-/- mice. In contrast, both CD8+ T-cell infiltration and proinflammatory/pro-tumour cytokines in the liver were significantly upregulated in Fah-/- mice during chronic liver injury, which was notably reversed by either metformin or atRA treatment. Regarding mechanisms, metformin regulated the decrease in Cyp26a1 enzyme expression and increased atRA expression via the AMPK/STAT3/Gadd45β/JNK/c-Jun pathway. CONCLUSIONS Metformin inhibits non-diabetic HCC by upregulating atRA levels and downregulating CD8+ T cells. This is the first reporting that the traditional drug metformin regulates the metabolite atRA via the Cyp26a1-involved pathway. The present study provides a potential application of metformin and atRA in non-diabetic HCC.
Collapse
Affiliation(s)
- Weizhi He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
- Fudan University Shanghai Cancer Center, International Co‐Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Shanghai Medical College of Fudan University, Institutes of Biomedical SciencesShanghai Key Laboratory of Medical EpigeneticsShanghaiChina
| | - Xicheng Wang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Miaomiao Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New AreaShanghaiChina
| | - Wenjian Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Lili Pan
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yangyang Cui
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoningChina
| | - Zhao Yu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yang Yang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Mingyang Xu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Zhaoxuan Dong
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Keming Ma
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic SurgeryShanghai East Hospital, Tongji UniversityShanghaiChina
| | - Zhiying He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| |
Collapse
|
38
|
Sunassee ED, Jardim-Perassi BV, Madonna MC, Ordway B, Ramanujam N. Metabolic Imaging as a Tool to Characterize Chemoresistance and Guide Therapy in Triple-Negative Breast Cancer (TNBC). Mol Cancer Res 2023; 21:995-1009. [PMID: 37343066 PMCID: PMC10592445 DOI: 10.1158/1541-7786.mcr-22-1004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
After an initial response to chemotherapy, tumor relapse is frequent. This event is reflective of both the spatiotemporal heterogeneities of the tumor microenvironment as well as the evolutionary propensity of cancer cell populations to adapt to variable conditions. Because the cause of this adaptation could be genetic or epigenetic, studying phenotypic properties such as tumor metabolism is useful as it reflects molecular, cellular, and tissue-level dynamics. In triple-negative breast cancer (TNBC), the characteristic metabolic phenotype is a highly fermentative state. However, during treatment, the spatial and temporal dynamics of the metabolic landscape are highly unstable, with surviving populations taking on a variety of metabolic states. Thus, longitudinally imaging tumor metabolism provides a promising approach to inform therapeutic strategies, and to monitor treatment responses to understand and mitigate recurrence. Here we summarize some examples of the metabolic plasticity reported in TNBC following chemotherapy and review the current metabolic imaging techniques available in monitoring chemotherapy responses clinically and preclinically. The ensemble of imaging technologies we describe has distinct attributes that make them uniquely suited for a particular length scale, biological model, and/or features that can be captured. We focus on TNBC to highlight the potential of each of these technological advances in understanding evolution-based therapeutic resistance.
Collapse
Affiliation(s)
- Enakshi D. Sunassee
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Megan C. Madonna
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bryce Ordway
- Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
39
|
Liu D, Wang Y, Li X, Wang Y, Zhang Z, Wang Z, Zhang X. Participation of protein metabolism in cancer progression and its potential targeting for the management of cancer. Amino Acids 2023; 55:1223-1246. [PMID: 37646877 DOI: 10.1007/s00726-023-03316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Cancer malignancies may broadly be described as heterogeneous disorders manifested by uncontrolled cellular growth/division and proliferation. Tumor cells utilize metabolic reprogramming to accomplish the upregulated nutritional requirements for sustaining their uncontrolled growth, proliferation, and survival. Metabolic reprogramming also called altered or dysregulated metabolism undergoes modification in normal metabolic pathways for anabolic precursor's generation that serves to continue biomass formation that sustains the growth, proliferation, and survival of carcinogenic cells under a nutrition-deprived microenvironment. A wide range of dysregulated/altered metabolic pathways encompassing different metabolic regulators have been described; however, the current review is focused to explain deeply the metabolic pathways modifications inducing upregulation of proteins/amino acids metabolism. The essential modification of various metabolic cycles with their consequent outcomes meanwhile explored promising therapeutic targets playing a pivotal role in metabolic regulation and is successfully employed for effective target-specific cancer treatment. The current review is aimed to understand the metabolic reprogramming of different proteins/amino acids involved in tumor progression along with potential therapeutic perspective elucidating targeted cancer therapy via these targets.
Collapse
Affiliation(s)
- Dalong Liu
- Department of Orthopedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yun Wang
- Department of Thoracic Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xiaojiang Li
- Department of Orthopedics, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yan Wang
- Department of Neurosurgery, People's Hospital of Jilin City, Jilin, 136200, China
| | - Zhiqiang Zhang
- Department of Orthopedics, Baishan Hospital of Traditional Chinese Medicine, Baishan, 134300, China
| | - Zhifeng Wang
- Department of Traditional Chinese Medicine, Changchun Chaoyang District Hospital of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xudong Zhang
- Department of Brain Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China.
| |
Collapse
|
40
|
Lim TKH, Skoulidis F, Kerr KM, Ahn MJ, Kapp JR, Soares FA, Yatabe Y. KRAS G12C in advanced NSCLC: Prevalence, co-mutations, and testing. Lung Cancer 2023; 184:107293. [PMID: 37683526 DOI: 10.1016/j.lungcan.2023.107293] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/15/2023] [Accepted: 07/05/2023] [Indexed: 09/10/2023]
Abstract
KRAS is the most commonly mutated oncogene in advanced, non-squamous, non-small cell lung cancer (NSCLC) in Western countries. Of the various KRAS mutants, KRAS G12C is the most common variant (~40%), representing 10-13% of advanced non-squamous NSCLC. Recent regulatory approvals of the KRASG12C-selective inhibitors sotorasib and adagrasib for patients with advanced or metastatic NSCLC harboring KRASG12C have transformed KRAS into a druggable target. In this review, we explore the evolving role of KRAS from a prognostic to a predictive biomarker in advanced NSCLC, discussing KRAS G12C biology, real-world prevalence, clinical relevance of co-mutations, and approaches to molecular testing. Real-world evidence demonstrates significant geographic differences in KRAS G12C prevalence (8.9-19.5% in the US, 9.3-18.4% in Europe, 6.9-9.0% in Latin America, and 1.4-4.3% in Asia) in advanced NSCLC. Additionally, the body of clinical data pertaining to KRAS G12C co-mutations such as STK11, KEAP1, and TP53 is increasing. In real-world evidence, KRAS G12C-mutant NSCLC was associated with STK11, KEAP1, and TP53 co-mutations in 10.3-28.0%, 6.3-23.0%, and 17.8-50.0% of patients, respectively. Whilst sotorasib and adagrasib are currently approved for use in the second-line setting and beyond for patients with advanced/metastatic NSCLC, testing and reporting of the KRAS G12C variant should be included in routine biomarker testing prior to first-line therapy. KRAS G12C test results should be clearly documented in patients' health records for actionability at progression. Where available, next-generation sequencing is recommended to facilitate simultaneous testing of potentially actionable biomarkers in a single run to conserve tissue. Results from molecular testing should inform clinical decisions in treating patients with KRAS G12C-mutated advanced NSCLC.
Collapse
Affiliation(s)
| | - Ferdinandos Skoulidis
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith M Kerr
- Department of Pathology, Aberdeen University Medical School and Aberdeen Royal Infirmary, Aberdeen, UK
| | - Myung-Ju Ahn
- Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | - Fernando A Soares
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil; Faculty of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan.
| |
Collapse
|
41
|
Kim JS, Kim MY, Hong S. Synergistic Effects of Metformin and Trastuzumab on HER2 Positive Gastroesophageal Adenocarcinoma Cells In Vitro and In Vivo. Cancers (Basel) 2023; 15:4768. [PMID: 37835462 PMCID: PMC10571931 DOI: 10.3390/cancers15194768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The incidence of HER2 amplification in advanced gastroesophageal adenocarcinoma (GC) reportedly ranges between 10% and 20%, depending on the population studied and the geographical region. Trastuzumab (Tmab) is the standard treatment for GCs with HER2 amplification. Metformin, a widely used antidiabetic drug, is an activator of AMP kinase that can affect the mTOR signaling pathway. The following GC cells were evaluated: HER2+ NCI-N87, YCC-19, YCC-38, OE19, OE33, and HER2- AGS. The effects of Tmab and metformin on these cell lines were assessed as single agents and in combination using cell viability assays, Western blotting, and xenograft models. Metformin induced phosphorylation of AMP kinase in all tested GC cells and dephosphorylation of mTOR in Tmab-sensitive GC cells. We observed that treatment with Tmab in combination with metformin induced a significant decrease in the number of colonies formed on soft agar by N87, YCC-19, YCC-38, and OE19 cells (88%, 95%, 73%, and 98%, respectively), in comparison to the number formed by control cells or cells in the single-treatment groups. No growth inhibition was detected in OE33 cells treated with Tmab alone. Combination with metformin resulted in decreased phosphorylation of HER2 and its downstream targets, AKT and ERK, in Tmab-sensitive HER2+ cells. Phospho-receptor tyrosine kinase (RTK) arrays were used to profile the phospho-proteome, which demonstrated a synergistic decrease in phosphorylation of EGFR, HER2, and HER3. Furthermore, the combination of Tmab and metformin exhibited enhanced antitumor effects in a xenograft model. Collectively, these data suggest that Tmab and metformin act synergistically in HER2+ GC cells. Since metformin is widely used and relatively non-toxic, its addition to the therapeutic regimen along with Tmab could enhance the clinical efficacy in patients with HER2+ GC.
Collapse
Affiliation(s)
- Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea;
| | - Mi Young Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea;
| | - Sungyoul Hong
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea;
| |
Collapse
|
42
|
Lu MZ, Li DY, Wang XF. Effect of metformin use on the risk and prognosis of ovarian cancer: an updated systematic review and meta-analysis. Panminerva Med 2023; 65:351-361. [PMID: 31290300 DOI: 10.23736/s0031-0808.19.03640-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Emerging evidence suggests that metformin has a potential antitumor effect both in vitro and in vivo. Increasing epidemiological studies indicate that diabetic patients receiving metformin therapy have lower incidences of cancer and have better survival rates. However, there are limited and inconsistent studies available about the effect of metformin therapy on ovarian cancer (OC). Thus, we conducted this meta-analysis to study the effect of metformin therapy on OC. Meanwhile, we systematically reviewed relevant studies to provide a framework for future research. EVIDENCE ACQUISITION We conducted a systematic literature search on PubMed, Web of Science, Springerlink, CNKI, VIP, SinoMed, and Wanfang up to the period of October 2018. A random-effects meta-analysis model was used to derive pooled effect estimates. EVIDENCE SYNTHESIS A total of 13 studies were retrieved of which 5 studies explained the prevention and 8 studies explained the treatment for OC. Our pooled results showed that metformin has a potential preventive effect on OC in diabetic women (pooled odds ratio [OR] 0.62, 95% confidence interval [95% CI] 0.34, 1.11; P<0.001). In addition, metformin can also significantly prolong progression-free survival (PFS) (pooled hazard ratio [HR] 0.49, 95% CI 0.34, 0.70; P=0.002), and overall survival (OS) (HR 0.71, 95%CI 0.61, 0.82; P<0.001) in patients with OC, regardless of whether they had diabetes. CONCLUSIONS The use of metformin can potentially reduce the risk of OC among diabetics, and it also can significantly improve PFS and OS in patients with OC. A further large clinical investigation would be needed to adopt our finding in practice, however, our systematic review provides an insight for future study designs.
Collapse
Affiliation(s)
- Min-Zhen Lu
- Second Clinical Medical College of Southern Medical University, Guangzhou, China -
| | - De-Yu Li
- Department of Oncology, Fujian Provincial Hospital, Fujian, China
| | - Xue-Feng Wang
- Department of Obstetrics and Gynecology, Third Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Pillai U J, Ray A, Maan M, Dutta M. Repurposing drugs targeting metabolic diseases for cancer therapeutics. Drug Discov Today 2023; 28:103684. [PMID: 37379903 DOI: 10.1016/j.drudis.2023.103684] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/01/2023] [Accepted: 06/18/2023] [Indexed: 06/30/2023]
Abstract
Hurdles in the identification of new drugs for cancer treatment have made drug repurposing an increasingly appealing alternative. The approach involves the use of old drugs for new therapeutic purposes. It is cost-effective and facilitates rapid clinical translation. Given that cancer is also considered a metabolic disease, drugs for metabolic disorders are being actively repurposed for cancer therapeutics. In this review, we discuss the repurposing of such drugs approved for two major metabolic diseases, diabetes and cardiovascular disease (CVD), which have shown potential as anti-cancer treatment. We also highlight the current understanding of the cancer signaling pathways that these drugs target.
Collapse
Affiliation(s)
- Jisha Pillai U
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Anindita Ray
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Meenu Maan
- Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, UAE; New York University-Abu Dhabi, Abu Dhabi, UAE.
| | - Mainak Dutta
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE.
| |
Collapse
|
44
|
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
|
45
|
Zhang H, Nabel CS, Li D, O'Connor RÍ, Crosby CR, Chang SM, Hao Y, Stanley R, Sahu S, Levin DS, Chen T, Tang S, Huang HY, Meynardie M, Stephens J, Sherman F, Chafitz A, Costelloe N, Rodrigues DA, Fogarty H, Kiernan MG, Cronin F, Papadopoulos E, Ploszaj M, Weerasekara V, Deng J, Kiely P, Bardeesy N, Vander Heiden MG, Chonghaile TN, Dowling CM, Wong KK. Histone Deacetylase 6 Inhibition Exploits Selective Metabolic Vulnerabilities in LKB1 Mutant, KRAS Driven NSCLC. J Thorac Oncol 2023; 18:882-895. [PMID: 36958689 PMCID: PMC10332301 DOI: 10.1016/j.jtho.2023.03.014] [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: 08/16/2022] [Revised: 01/24/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
INTRODUCTION In KRAS-mutant NSCLC, co-occurring alterations in LKB1 confer a negative prognosis compared with other mutations such as TP53. LKB1 is a tumor suppressor that coordinates several signaling pathways in response to energetic stress. Our recent work on pharmacologic and genetic inhibition of histone deacetylase 6 (HDAC6) revealed the impaired activity of numerous enzymes involved in glycolysis. On the basis of these previous findings, we explored the therapeutic window for HDAC6 inhibition in metabolically-active KRAS-mutant lung tumors. METHODS Using cell lines derived from mouse autochthonous tumors bearing the KRAS/LKB1 (KL) and KRAS/TP53 mutant genotypes to control for confounding germline and somatic mutations in human models, we characterize the metabolic phenotypes at baseline and in response to HDAC6 inhibition. The impact of HDAC6 inhibition was measured on cancer cell growth in vitro and on tumor growth in vivo. RESULTS Surprisingly, KL-mutant cells revealed reduced levels of redox-sensitive cofactors at baseline. This is associated with increased sensitivity to pharmacologic HDAC6 inhibition with ACY-1215 and blunted ability to increase compensatory metabolism and buffer oxidative stress. Seeking synergistic metabolic combination treatments, we found enhanced cell killing and antitumor efficacy with glutaminase inhibition in KL lung cancer models in vitro and in vivo. CONCLUSIONS Exploring the differential metabolism of KL and KRAS/TP53-mutant NSCLC, we identified decreased metabolic reserve in KL-mutant tumors. HDAC6 inhibition exploited a therapeutic window in KL NSCLC on the basis of a diminished ability to compensate for impaired glycolysis, nominating a novel strategy for the treatment of KRAS-mutant NSCLC with co-occurring LKB1 mutations.
Collapse
Affiliation(s)
- Hua Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania; Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Christopher S Nabel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts; Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dezhi Li
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Ruth Í O'Connor
- School of Medicine, University of Limerick, Limerick, Ireland
| | - Caroline R Crosby
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sarah M Chang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, New York
| | - Robyn Stanley
- School of Medicine, University of Limerick, Limerick, Ireland
| | - Soumyadip Sahu
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Daniel S Levin
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Ting Chen
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Sittinon Tang
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Hsin-Yi Huang
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Mary Meynardie
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Janaye Stephens
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Fiona Sherman
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Alison Chafitz
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | | | - Daniel A Rodrigues
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hilda Fogarty
- School of Medicine, University of Limerick, Limerick, Ireland
| | | | - Fiona Cronin
- School of Medicine, University of Limerick, Limerick, Ireland
| | - Eleni Papadopoulos
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Magdalena Ploszaj
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Vajira Weerasekara
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jiehui Deng
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Patrick Kiely
- School of Medicine, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts; Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Triona Ni Chonghaile
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Catríona M Dowling
- School of Medicine, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland.
| | - Kwok-Kin Wong
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| |
Collapse
|
46
|
Tan I, Xu S, Huo J, Huang Y, Lim HH, Lam KP. Identification of a novel mitochondria-localized LKB1 variant required for the regulation of the oxidative stress response. J Biol Chem 2023; 299:104906. [PMID: 37302555 PMCID: PMC10404683 DOI: 10.1016/j.jbc.2023.104906] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023] Open
Abstract
The tumor suppressor Liver Kinase B1 (LKB1) is a multifunctional serine/threonine protein kinase that regulates cell metabolism, polarity, and growth and is associated with Peutz-Jeghers Syndrome and cancer predisposition. The LKB1 gene comprises 10 exons and 9 introns. Three spliced LKB1 variants have been documented, and they reside mainly in the cytoplasm, although two possess a nuclear-localization sequence (NLS) and are able to shuttle into the nucleus. Here, we report the identification of a fourth and novel LKB1 isoform that is, interestingly, targeted to the mitochondria. We show that this mitochondria-localized LKB1 (mLKB1) is generated from alternative splicing in the 5' region of the transcript and translated from an alternative initiation codon encoded by a previously unknown exon 1b (131 bp) hidden within the long intron 1 of LKB1 gene. We found by replacing the N-terminal NLS of the canonical LKB1 isoform, the N-terminus of the alternatively spliced mLKB1 variant encodes a mitochondrial transit peptide that allows it to localize to the mitochondria. We further demonstrate that mLKB1 colocalizes histologically with mitochondria-resident ATP Synthase and NAD-dependent deacetylase sirtuin-3, mitochondrial (SIRT3) and that its expression is rapidly and transiently upregulated by oxidative stress. We conclude that this novel LKB1 isoform, mLKB1, plays a critical role in regulating mitochondrial metabolic activity and oxidative stress response.
Collapse
Affiliation(s)
- Ivan Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianxin Huo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yuhan Huang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Hong-Hwa Lim
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
47
|
Pillai R, LeBoeuf SE, Hao Y, New C, Blum JLE, Rashidfarrokhi A, Huang SM, Bahamon C, Wu WL, Karadal-Ferrena B, Herrera A, Ivanova E, Cross M, Bossowski JP, Ding H, Hayashi M, Rajalingam S, Karakousi T, Sayin VI, Khanna KM, Wong KK, Wild R, Tsirigos A, Poirier JT, Rudin CM, Davidson SM, Koralov SB, Papagiannakopoulos T. Glutamine antagonist DRP-104 suppresses tumor growth and enhances response to checkpoint blockade in KEAP1 mutant lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546750. [PMID: 37425844 PMCID: PMC10327154 DOI: 10.1101/2023.06.27.546750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Loss-of-function mutations in KEAP1 frequently occur in lung cancer and are associated with resistance to standard of care treatment, highlighting the need for the development of targeted therapies. We have previously shown that KEAP1 mutant tumors have increased glutamine consumption to support the metabolic rewiring associated with NRF2 activation. Here, using patient-derived xenograft models and antigenic orthotopic lung cancer models, we show that the novel glutamine antagonist DRP-104 impairs the growth of KEAP1 mutant tumors. We find that DRP-104 suppresses KEAP1 mutant tumor growth by inhibiting glutamine-dependent nucleotide synthesis and promoting anti-tumor CD4 and CD8 T cell responses. Using multimodal single-cell sequencing and ex vivo functional assays, we discover that DRP-104 reverses T cell exhaustion and enhances the function of CD4 and CD8 T cells culminating in an improved response to anti-PD1 therapy. Our pre-clinical findings provide compelling evidence that DRP-104, currently in phase 1 clinical trials, offers a promising therapeutic approach for treating patients with KEAP1 mutant lung cancer. Furthermore, we demonstrate that by combining DRP-104 with checkpoint inhibition, we can achieve suppression of tumor intrinsic metabolism and augmentation of anti-tumor T cell responses.
Collapse
|
48
|
Verma S, Chitikela S, Singh V, Khurana S, Pushpam D, Jain D, Kumar S, Gupta Y, Malik PS. A phase II study of metformin plus pemetrexed and carboplatin in patients with non-squamous non-small cell lung cancer (METALUNG). Med Oncol 2023; 40:192. [PMID: 37261532 DOI: 10.1007/s12032-023-02057-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
Immune checkpoint inhibitors (ICIs) ± chemotherapy is the standard treatment for driver mutation-negative non-small cell lung cancer (NSCLC). However, accessibility to ICIs in LMICs is limited due to high cost, and platinum-based chemotherapy remains the mainstay of treatment. Metformin has anticancer properties, and studies suggest synergism between metformin and pemetrexed. Based on preclinical evidence, this combination may be more beneficial for STK11-mutated NSCLC, a subgroup, inherently resistant to ICIs. In this Simon two-stage, single-arm phase 2 trial, we investigated metformin with pemetrexed-carboplatin (PC) in patients with treatment-naive stage IV non-squamous NSCLC. The primary outcome was 6-month progression-free survival (PFS) rate. Secondary outcomes were safety, overall survival (OS), overall response rate (ORR), proportion of STK11 mutation, and effect of STK11 mutation on 6-month PFS rate. The study was terminated for futility after interim analysis. The median follow-up was 34.1 months. The 6-month PFS rate was 28% (95% CI 12.4-0.46). The median PFS and OS were 4.5 (95% CI 2.2-6.1) and 7.4 months (95% CI 5.3-15.3), respectively. The ORR was 72%. Gastrointestinal toxicities were the most common. No grade 4/5 toxicities were reported. Targeted sequencing was possible in nine cases. Two patients had STK11 mutation and a poor outcome (PFS < 12 weeks). We could not demonstrate the benefit of metformin with CP in terms of improvement in 6-month PFS rate; however, the combination was safe (CTRI/2019/02/017815).
Collapse
Affiliation(s)
- S Verma
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - S Chitikela
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - V Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - S Khurana
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - D Pushpam
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - D Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - S Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Y Gupta
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - P S Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India.
- Department of Medical Oncology, Dr.B.R.A.I.R.C.H., All India Institute of Medical Sciences, Room 245, New Delhi, India.
| |
Collapse
|
49
|
Hsu BW, Chen BS. Genetic and Epigenetic Host-Virus Network to Investigate Pathogenesis and Identify Biomarkers for Drug Repurposing of Human Respiratory Syncytial Virus via Real-World Two-Side RNA-Seq Data: Systems Biology and Deep-Learning Approach. Biomedicines 2023; 11:1531. [PMID: 37371627 DOI: 10.3390/biomedicines11061531] [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: 03/28/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) affects more than 33 million people each year, but there are currently no effective drugs or vaccines approved. In this study, we first constructed a candidate host-pathogen interspecies genome-wide genetic and epigenetic network (HPI-GWGEN) via big-data mining. Then, we employed reversed dynamic methods via two-side host-pathogen RNA-seq time-profile data to prune false positives in candidate HPI-GWGEN to obtain the real HPI-GWGEN. With the aid of principal-network projection and the annotation of KEGG pathways, we can extract core signaling pathways during hRSV infection to investigate the pathogenic mechanism of hRSV infection and select the corresponding significant biomarkers as drug targets, i.e., TRAF6, STAT3, IRF3, TYK2, and MAVS. Finally, in order to discover potential molecular drugs, we trained a DNN-based DTI model by drug-target interaction databases to predict candidate molecular drugs for these drug targets. After screening these candidate molecular drugs by three drug design specifications simultaneously, i.e., regulation ability, sensitivity, and toxicity. We finally selected acitretin, RS-67333, and phenformin to combine as a potential multimolecule drug for the therapeutic treatment of hRSV infection.
Collapse
Affiliation(s)
- Bo-Wei Hsu
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Bor-Sen Chen
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
50
|
Compton SE, Kitchen-Goosen SM, DeCamp LM, Lau KH, Mabvakure B, Vos M, Williams KS, Wong KK, Shi X, Rothbart SB, Krawczyk CM, Jones RG. LKB1 controls inflammatory potential through CRTC2-dependent histone acetylation. Mol Cell 2023:S1097-2765(23)00288-5. [PMID: 37172591 DOI: 10.1016/j.molcel.2023.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential.
Collapse
Affiliation(s)
- Shelby E Compton
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Susan M Kitchen-Goosen
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kin H Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Batsirai Mabvakure
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Matthew Vos
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Scott B Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Connie M Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|