51
|
Wang H, Hu Q, Chen Y, Huang X, Feng Y, Shi Y, Li R, Yin X, Song X, Liang Y, Zhang T, Xu L, Dong G, Jiang F. Ferritinophagy mediates adaptive resistance to EGFR tyrosine kinase inhibitors in non-small cell lung cancer. Nat Commun 2024; 15:4195. [PMID: 38760351 PMCID: PMC11101634 DOI: 10.1038/s41467-024-48433-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/26/2024] [Indexed: 05/19/2024] Open
Abstract
Osimertinib (Osi) is a widely used epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). However, the emergence of resistance is inevitable, partly due to the gradual evolution of adaptive resistant cells during initial treatment. Here, we find that Osi treatment rapidly triggers adaptive resistance in tumor cells. Metabolomics analysis reveals a significant enhancement of oxidative phosphorylation (OXPHOS) in Osi adaptive-resistant cells. Mechanically, Osi treatment induces an elevation of NCOA4, a key protein of ferritinophagy, which maintains the synthesis of iron-sulfur cluster (ISC) proteins of electron transport chain and OXPHOS. Additionally, active ISC protein synthesis in adaptive-resistant cells significantly increases the sensitivity to copper ions. Combining Osi with elesclomol, a copper ion ionophore, significantly increases the efficacy of Osi, with no additional toxicity. Altogether, this study reveals the mechanisms of NCOA4-mediated ferritinophagy in Osi adaptive resistance and introduces a promising new therapy of combining copper ionophores to improve its initial efficacy.
Collapse
Affiliation(s)
- Hui Wang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Qianfan Hu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yuzhong Chen
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Xing Huang
- Department of Pathology, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Yipeng Feng
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yuanjian Shi
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Rutao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuewen Yin
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
| | - Xuming Song
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yingkuan Liang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
| | - Te Zhang
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Lin Xu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Jiangning District, Nanjing, China
| | - Gaochao Dong
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China.
| | - Feng Jiang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Xuanwu District, Nanjing, China.
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Xuanwu District, Nanjing, China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
52
|
Zhou H, Li G, Kan L, Yang M, Liu Y, Miu X, Shi L, Yang Z, Zheng X, Chen H, Ren C. Synergistic induction of autophagy in gastric cancer by targeting CDK4/6 and MEK through AMPK/mTOR pathway. Heliyon 2024; 10:e30475. [PMID: 38726124 PMCID: PMC11079098 DOI: 10.1016/j.heliyon.2024.e30475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
KRAS is a commonly mutated oncogene in human gastric cancer and is often associated with drug resistance and poor prognosis. Co-clinical trial of combined MEK-CDK4/6 inhibition in KRAS mutated cancers demonstrated therapeutic efficacy in patient-derived xenografts and safety in patients. Here, present research focuses on targeting CDK4/6 and MEK synergistically block the proliferation of KRAS-mutated gastric cancer cells in vitro and in vivo and induced autophagy through the AMPK/mTOR pathway. Furthermore, autophagy inhibitor combined with targeting CDK4/6 and MEK therapy had significant antitumor effects on KRAS mutant gastric cancer cells. Clinical trials are needed to determine the mechanism behind this finding and its clinical utility. In conclusion, our results demonstrate autophagy inhibitor combined targeting MEK and CDK4/6 that concurrently block multiple metabolic processes may be an effective therapeutic approach for gastric cancer.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Guiling Li
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Liuyue Kan
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Mingyu Yang
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Yu Liu
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Xiaye Miu
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
| | - Lei Shi
- Department of Gastrointestinal Surgery, Clinical College of Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Zhanjun Yang
- Department of Chemistry, Yangzhou University, Yangzhou, Jiangsu, 225002, China
| | - Xucai Zheng
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Hui Chen
- Department of Geriatrics, Clinical College of Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Chuanli Ren
- Department of Laboratory Medicine, Clinical College of Yangzhou University and Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, China
- Department of Laboratory Medicine, Clinical College of Xuzhou Medica University, Yangzhou, Jiangsu, 225001, China
| |
Collapse
|
53
|
Perurena N, Situ L, Cichowski K. Combinatorial strategies to target RAS-driven cancers. Nat Rev Cancer 2024; 24:316-337. [PMID: 38627557 DOI: 10.1038/s41568-024-00679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 05/01/2024]
Abstract
Although RAS was formerly considered undruggable, various agents that inhibit RAS or specific RAS oncoproteins have now been developed. Indeed, the importance of directly targeting RAS has recently been illustrated by the clinical success of mutant-selective KRAS inhibitors. Nevertheless, responses to these agents are typically incomplete and restricted to a subset of patients, highlighting the need to develop more effective treatments, which will likely require a combinatorial approach. Vertical strategies that target multiple nodes within the RAS pathway to achieve deeper suppression are being investigated and have precedence in other contexts. However, alternative strategies that co-target RAS and other therapeutic vulnerabilities have been identified, which may mitigate the requirement for profound pathway suppression. Regardless, the efficacy of any given approach will likely be dictated by genetic, epigenetic and tumour-specific variables. Here we discuss various combinatorial strategies to treat KRAS-driven cancers, highlighting mechanistic concepts that may extend to tumours harbouring other RAS mutations. Although many promising combinations have been identified, clinical responses will ultimately depend on whether a therapeutic window can be achieved and our ability to prospectively select responsive patients. Therefore, we must continue to develop and understand biologically diverse strategies to maximize our likelihood of success.
Collapse
Affiliation(s)
- Naiara Perurena
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lisa Situ
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Ludwig Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
54
|
Cheng K, Zhou Z, Chen Q, Chen Z, Cai Y, Cai H, Wu S, Gao P, Cai Y, Zhou J, Wang X, Wu Z, Peng B. CDK4/6 inhibition sensitizes MEK inhibition by inhibiting cell cycle and proliferation in pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:8389. [PMID: 38600093 PMCID: PMC11006845 DOI: 10.1038/s41598-024-57417-z] [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: 11/22/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is not sensitive to most chemotherapy drugs, leading to poor chemotherapy efficacy. Recently, Trametinib and Palbociclib have promising prospects in the treatment of pancreatic cancer. This article aims to explore the effects of Trametinib on pancreatic cancer and address the underlying mechanism of resistance as well as its reversal strategies. The GDSC (Genomics of Drug Sensitivity in Cancer) and CTD2 (Cancer Target Discovery and Development) were utilized to screen the potential drug candidate in PDAC cell lines. The dose-increase method combined with the high-dose shock method was applied to induce the Trametinib-resistant PANC-1 and MIA PaCa-2 cell lines. The CCK8 proliferation assay, colony formation assay, flow cytometry, and western blot were conducted to verify the inhibitory effect of Trametinib and Palbociclib. RNA-seq was performed in resistant PDAC cell lines to find the differential expression genes related to drug resistance and predict pathways leading to the reversal of Trametinib resistance. The GDSC and CTD2 database screening revealed that Trametinib demonstrates a significant inhibitory effect on PDAC. We found that Trametinib has a lower IC50 than Gemcitabine in PDAC cell lines. Both Trametinib and Gemcitabine can decrease the proliferation capacity of pancreatic cells, induce cell cycle arrest, and increase apoptosis. Simultaneously, the phosphorylation of the AKT and ERK pathways were inhibited by the treatment of Trametinib. In addition, the RNA-seq of Trametinib-induced resistance PDAC cell lines reveals that the cyclin-dependent kinase (CDK)-RB-E2F regulatory axis and G2/M DNA damage checkpoint might lead the drug resistance. Besides, the combination of Trametinib with Palbociclib could inhibit the proliferation and cell cycle of both resistant cells lines and also restore the sensitivity of drug-resistant cells to Trametinib. Last but not least, the interferon-α and interferon-γ expression were upregulated in resistance cell lines, which might lead to the reversal of drug resistance. The study shows Trametinib has a critical inhibitory effect on PDAC. Besides, the combination of Trametinib with Palbociclib can inhibit the proliferation of PDAC-resistant cells.
Collapse
Affiliation(s)
- Ke Cheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zijian Zhou
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiangxing Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zixin Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yu Cai
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - He Cai
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shangdi Wu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Pan Gao
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yunqiang Cai
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Zhou
- Division of Liver Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xin Wang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhong Wu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China.
| | - Bing Peng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
55
|
Schmid M, Fischer P, Engl M, Widder J, Kerschbaum-Gruber S, Slade D. The interplay between autophagy and cGAS-STING signaling and its implications for cancer. Front Immunol 2024; 15:1356369. [PMID: 38660307 PMCID: PMC11039819 DOI: 10.3389/fimmu.2024.1356369] [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: 12/15/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Autophagy is an intracellular process that targets various cargos for degradation, including members of the cGAS-STING signaling cascade. cGAS-STING senses cytosolic double-stranded DNA and triggers an innate immune response through type I interferons. Emerging evidence suggests that autophagy plays a crucial role in regulating and fine-tuning cGAS-STING signaling. Reciprocally, cGAS-STING pathway members can actively induce canonical as well as various non-canonical forms of autophagy, establishing a regulatory network of feedback mechanisms that alter both the cGAS-STING and the autophagic pathway. The crosstalk between autophagy and the cGAS-STING pathway impacts a wide variety of cellular processes such as protection against pathogenic infections as well as signaling in neurodegenerative disease, autoinflammatory disease and cancer. Here we provide a comprehensive overview of the mechanisms involved in autophagy and cGAS-STING signaling, with a specific focus on the interactions between the two pathways and their importance for cancer.
Collapse
Affiliation(s)
- Maximilian Schmid
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| | - Patrick Fischer
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| | - Magdalena Engl
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
- Vienna Biocenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Joachim Widder
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sylvia Kerschbaum-Gruber
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
| | - Dea Slade
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- MedAustron Ion Therapy Center, Wiener Neustadt, Austria
- Department of Medical Biochemistry, Medical University of Vienna, Max Perutz Labs, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
56
|
Singhal A, Li BT, O'Reilly EM. Targeting KRAS in cancer. Nat Med 2024; 30:969-983. [PMID: 38637634 PMCID: PMC11845254 DOI: 10.1038/s41591-024-02903-0] [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: 01/04/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024]
Abstract
RAS family variants-most of which involve KRAS-are the most commonly occurring hotspot mutations in human cancers and are associated with a poor prognosis. For almost four decades, KRAS has been considered undruggable, in part due to its structure, which lacks small-molecule binding sites. But recent developments in bioengineering, organic chemistry and related fields have provided the infrastructure to make direct KRAS targeting possible. The first successes occurred with allele-specific targeting of KRAS p.Gly12Cys (G12C) in non-small cell lung cancer, resulting in regulatory approval of two agents-sotorasib and adagrasib. Inhibitors targeting other variants beyond G12C have shown preliminary antitumor activity in highly refractory malignancies such as pancreatic cancer. Herein, we outline RAS pathobiology with a focus on KRAS, illustrate therapeutic approaches across a variety of malignancies, including emphasis on the 'on' and 'off' switch allele-specific and 'pan' RAS inhibitors, and review immunotherapeutic and other key combination RAS targeting strategies. We summarize mechanistic understanding of de novo and acquired resistance, review combination approaches, emerging technologies and drug development paradigms and outline a blueprint for the future of KRAS therapeutics with anticipated profound clinical impact.
Collapse
Affiliation(s)
- Anupriya Singhal
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bob T Li
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
57
|
Cortesi A, Gandolfi F, Arco F, Di Chiaro P, Valli E, Polletti S, Noberini R, Gualdrini F, Attanasio S, Citron F, Ho IL, Shah R, Yen EY, Spinella MC, Ronzoni S, Rodighiero S, Mitro N, Bonaldi T, Ghisletti S, Monticelli S, Viale A, Diaferia GR, Natoli G. Activation of endogenous retroviruses and induction of viral mimicry by MEK1/2 inhibition in pancreatic cancer. SCIENCE ADVANCES 2024; 10:eadk5386. [PMID: 38536927 PMCID: PMC10971493 DOI: 10.1126/sciadv.adk5386] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/21/2024] [Indexed: 02/08/2025]
Abstract
While pancreatic ductal adenocarcinomas (PDACs) are addicted to KRAS-activating mutations, inhibitors of downstream KRAS effectors, such as the MEK1/2 kinase inhibitor trametinib, are devoid of therapeutic effects. However, the extensive rewiring of regulatory circuits driven by the attenuation of the KRAS pathway may induce vulnerabilities of therapeutic relevance. An in-depth molecular analysis of the transcriptional and epigenomic alterations occurring in PDAC cells in the initial hours after MEK1/2 inhibition by trametinib unveiled the induction of endogenous retroviruses (ERVs) escaping epigenetic silencing, leading to the production of double-stranded RNAs and the increased expression of interferon (IFN) genes. We tracked ERV activation to the early induction of the transcription factor ELF3, which extensively bound and activated nonsilenced retroelements and synergized with IRF1 (interferon regulatory factor 1) in the activation of IFNs and IFN-stimulated genes. Trametinib-induced viral mimicry in PDAC may be exploited in the rational design of combination therapies in immuno-oncology.
Collapse
Affiliation(s)
- Alice Cortesi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Francesco Gandolfi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Fabiana Arco
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Pierluigi Di Chiaro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Emanuele Valli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Sara Polletti
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Roberta Noberini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Francesco Gualdrini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Sergio Attanasio
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - I-lin Ho
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rutvi Shah
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Er-Yen Yen
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mara Cetty Spinella
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Simona Ronzoni
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Nico Mitro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti,” Università degli Studi di Milano, Milano 20133, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti,” Università degli Studi di Milano, Milano 20133, Italy
| | - Serena Ghisletti
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Silvia Monticelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Gioacchino Natoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| |
Collapse
|
58
|
Bulle A, Liu P, Seehra K, Bansod S, Chen Y, Zahra K, Somani V, Khawar IA, Chen HP, Dodhiawala PB, Li L, Geng Y, Mo CK, Mahsl J, Ding L, Govindan R, Davies S, Mudd J, Hawkins WG, Fields RC, DeNardo DG, Knoerzer D, Held JM, Grierson PM, Wang-Gillam A, Ruzinova MB, Lim KH. Combined KRAS-MAPK pathway inhibitors and HER2-directed drug conjugate is efficacious in pancreatic cancer. Nat Commun 2024; 15:2503. [PMID: 38509064 PMCID: PMC10954758 DOI: 10.1038/s41467-024-46811-w] [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: 08/02/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Targeting the mitogen-activated protein kinase (MAPK) cascade in pancreatic ductal adenocarcinoma (PDAC) remains clinically unsuccessful. We aim to develop a MAPK inhibitor-based therapeutic combination with strong preclinical efficacy. Utilizing a reverse-phase protein array, we observe rapid phospho-activation of human epidermal growth factor receptor 2 (HER2) in PDAC cells upon pharmacological MAPK inhibition. Mechanistically, MAPK inhibitors lead to swift proteasomal degradation of dual-specificity phosphatase 6 (DUSP6). The carboxy terminus of HER2, containing a TEY motif also present in extracellular signal-regulated kinase 1/2 (ERK1/2), facilitates binding with DUSP6, enhancing its phosphatase activity to dephosphorylate HER2. In the presence of MAPK inhibitors, DUSP6 dissociates from the protective effect of the RING E3 ligase tripartite motif containing 21, resulting in its degradation. In PDAC patient-derived xenograft (PDX) models, combining ERK and HER inhibitors slows tumour growth and requires cytotoxic chemotherapy to achieve tumour regression. Alternatively, MAPK inhibitors with trastuzumab deruxtecan, an anti-HER2 antibody conjugated with cytotoxic chemotherapy, lead to sustained tumour regression in most tested PDXs without causing noticeable toxicity. Additionally, KRAS inhibitors also activate HER2, supporting testing the combination of KRAS inhibitors and trastuzumab deruxtecan in PDAC. This study identifies a rational and promising therapeutic combination for clinical testing in PDAC patients.
Collapse
Affiliation(s)
- Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Peng Liu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kuljeet Seehra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yali Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kiran Zahra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vikas Somani
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Iftikhar Ali Khawar
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hung-Po Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yutong Geng
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chia-Kuei Mo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jay Mahsl
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Li Ding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ramaswamy Govindan
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sherri Davies
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jacqueline Mudd
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - William G Hawkins
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan C Fields
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David G DeNardo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Jason M Held
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
59
|
Cheng C, Hu J, Mannan R, Bhattacharyya R, Rossiter NJ, Magnuson B, Wisniewski JP, Zheng Y, Xiao L, Li C, Awad D, He T, Bao Y, Zhang Y, Cao X, Wang Z, Mehra R, Morlacchi P, Sahai V, di Magliano MP, Shah YM, Ding K, Qiao Y, Lyssiotis CA, Chinnaiyan AM. Targeting PIKfyve-driven lipid homeostasis as a metabolic vulnerability in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585580. [PMID: 38562800 PMCID: PMC10983929 DOI: 10.1101/2024.03.18.585580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) subsists in a nutrient-deregulated microenvironment, making it particularly susceptible to treatments that interfere with cancer metabolism12. For example, PDAC utilizes and is dependent on high levels of autophagy and other lysosomal processes3-5. Although targeting these pathways has shown potential in preclinical studies, progress has been hampered by the challenge of identifying and characterizing favorable targets for drug development6. Here, we characterize PIKfyve, a lipid kinase integral to lysosomal functioning7, as a novel and targetable vulnerability in PDAC. In human patient and murine PDAC samples, we discovered that PIKFYVE is overexpressed in PDAC cells compared to adjacent normal cells. Employing a genetically engineered mouse model, we established the essential role of PIKfyve in PDAC progression. Further, through comprehensive metabolic analyses, we found that PIKfyve inhibition obligated PDAC to upregulate de novo lipid synthesis, a relationship previously undescribed. PIKfyve inhibition triggered a distinct lipogenic gene expression and metabolic program, creating a dependency on de novo lipid metabolism pathways, by upregulating genes such as FASN and ACACA. In PDAC, the KRAS-MAPK signaling pathway is a primary driver of de novo lipid synthesis, specifically enhancing FASN and ACACA levels. Accordingly, the simultaneous targeting of PIKfyve and KRAS-MAPK resulted in the elimination of tumor burden in a syngeneic orthotopic model and tumor regression in a xenograft model of PDAC. Taken together, these studies suggest that disrupting lipid metabolism through PIKfyve inhibition induces synthetic lethality in conjunction with KRAS-MAPK-directed therapies for PDAC.
Collapse
Affiliation(s)
- Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PRC
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rupam Bhattacharyya
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas J Rossiter
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Brian Magnuson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jasmine P Wisniewski
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Chungen Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, PRC
| | - Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Vaibhav Sahai
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marina Pasca di Magliano
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PRC
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
60
|
Sun D, Zhang Z, Yu X, Li H, Wang X, Chen L. The mechanism of UNC-51-like kinase 1 and the applications of small molecule modulators in cancer treatment. Eur J Med Chem 2024; 268:116273. [PMID: 38432059 DOI: 10.1016/j.ejmech.2024.116273] [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: 12/29/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Autophagy is a process of self-renewal in cells, which not only provides the necessary nutrients for cells, but also clears necrotic organelles. Autophagy disorders are closely related to diseases such as cancer. UNC-51-like kinase 1 (ULK1) is a serine/threonine protein kinase that plays a crucial role in receiving input from energy and nutrient sensors, activating autophagy to maintain cellular homeostasis under stressful conditions. In recent years, targeting ULK1 has become a highly promising strategy for cancer treatment. This review introduces the regulatory mechanism of ULK1 in autophagy through the AMPK/mTOR/ULK1 pathway and reviews the research progress of ULK1 activators and inhibitors and their applications in cancer treatment. In addition, we analyze the binding modes between ULK1 and modulators through virtual molecular docking, which will provide a reliable basis and theoretical guidance for the design and development of new therapeutic drugs targeting ULK1.
Collapse
Affiliation(s)
- Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Chinese People's Liberation Army Logistics Support Force, No. 967 Hospital, Dalian, 116021, China
| | - Zhiqi Zhang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xinbo Yu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Xiaobo Wang
- Chinese People's Liberation Army Logistics Support Force, No. 967 Hospital, Dalian, 116021, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
61
|
Liu Z, Lu T, Qian R, Wang Z, Qi R, Zhang Z. Exploiting Nanotechnology for Drug Delivery: Advancing the Anti-Cancer Effects of Autophagy-Modulating Compounds in Traditional Chinese Medicine. Int J Nanomedicine 2024; 19:2507-2528. [PMID: 38495752 PMCID: PMC10944250 DOI: 10.2147/ijn.s455407] [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: 12/17/2023] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background Cancer continues to be a prominent issue in the field of medicine, as demonstrated by recent studies emphasizing the significant role of autophagy in the development of cancer. Traditional Chinese Medicine (TCM) provides a variety of anti-tumor agents capable of regulating autophagy. However, the clinical application of autophagy-modulating compounds derived from TCM is impeded by their restricted water solubility and bioavailability. To overcome this challenge, the utilization of nanotechnology has been suggested as a potential solution. Nonetheless, the current body of literature on nanoparticles delivering TCM-derived autophagy-modulating anti-tumor compounds for cancer treatment is limited, lacking comprehensive summaries and detailed descriptions. Methods Up to November 2023, a comprehensive research study was conducted to gather relevant data using a variety of databases, including PubMed, ScienceDirect, Springer Link, Web of Science, and CNKI. The keywords utilized in this investigation included "autophagy", "nanoparticles", "traditional Chinese medicine" and "anticancer". Results This review provides a comprehensive analysis of the potential of nanotechnology in overcoming delivery challenges and enhancing the anti-cancer properties of autophagy-modulating compounds in TCM. The evaluation is based on a synthesis of different classes of autophagy-modulating compounds in TCM, their mechanisms of action in cancer treatment, and their potential benefits as reported in various scholarly sources. The findings indicate that nanotechnology shows potential in enhancing the availability of autophagy-modulating agents in TCM, thereby opening up a plethora of potential therapeutic avenues. Conclusion Nanotechnology has the potential to enhance the anti-tumor efficacy of autophagy-modulating compounds in traditional TCM, through regulation of autophagy.
Collapse
Affiliation(s)
- Zixian Liu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Tianming Lu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruoning Qian
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zian Wang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruogu Qi
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zhengguang Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
62
|
Molina-Arcas M, Downward J. Exploiting the therapeutic implications of KRAS inhibition on tumor immunity. Cancer Cell 2024; 42:338-357. [PMID: 38471457 DOI: 10.1016/j.ccell.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
Over the past decade, RAS oncogenic proteins have transitioned from being deemed undruggable to having two clinically approved drugs, with several more in advanced stages of development. Despite the initial benefit of KRAS-G12C inhibitors for patients with tumors harboring this mutation, the rapid emergence of drug resistance underscores the urgent need to synergize these inhibitors with other therapeutic approaches to improve outcomes. RAS mutant tumor cells can create an immunosuppressive tumor microenvironment (TME), suggesting an increased susceptibility to immunotherapies following RAS inhibition. This provides a rationale for combining RAS inhibitory drugs with immune checkpoint blockade (ICB). However, achieving this synergy in the clinical setting has proven challenging. Here, we explore how understanding the impact of RAS mutant tumor cells on the TME can guide innovative approaches to combining RAS inhibition with immunotherapies, review progress in both pre-clinical and clinical stages, and discuss challenges and future directions.
Collapse
Affiliation(s)
| | - Julian Downward
- Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
63
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
64
|
Bildik G, Gray JP, Mao W, Yang H, Ozyurt R, Orellana VR, De Wever O, Carey MS, Bast RC, Lu Z. DIRAS3 induces autophagy and enhances sensitivity to anti-autophagic therapy in KRAS-driven pancreatic and ovarian carcinomas. Autophagy 2024; 20:675-691. [PMID: 38169324 PMCID: PMC10936598 DOI: 10.1080/15548627.2023.2299516] [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: 04/12/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) and low-grade ovarian cancer (LGSOC) are characterized by the prevalence of KRAS oncogene mutations. DIRAS3 is the first endogenous non-RAS protein that heterodimerizes with RAS, disrupts RAS clustering, blocks RAS signaling, and inhibits cancer cell growth. Here, we found that DIRAS3-mediated KRAS inhibition induces ROS-mediated apoptosis in PDAC and LGSOC cells with KRAS mutations, but not in cells with wild-type KRAS, by downregulating NFE2L2/Nrf2 transcription, reducing antioxidants, and inducing oxidative stress. DIRAS3 also induces cytoprotective macroautophagy/autophagy that may protect mutant KRAS cancer cells from oxidative stress, by inhibiting mutant KRAS, activating the STK11/LKB1-PRKAA/AMPK pathway, increasing lysosomal CDKN1B/p27 localization, and inducing autophagic gene expression. Treatment with chloroquine or the novel dimeric chloroquine analog DC661 significantly enhances DIRAS3-mediated inhibition of mutant KRAS tumor cell growth in vitro and in vivo. Taken together, our study demonstrates that DIRAS3 plays a critical role in regulating mutant KRAS-driven oncogenesis in PDAC and LGSOC.Abbreviations: AFR: autophagic flux reporter; ATG: autophagy related; CQ: chloroquine; DCFDA: 2'-7'-dichlorodihydrofluorescein diacetate; DIRAS3: DIRAS family GTPase 3; DOX: doxycycline; KRAS: KRAS proto-oncogene, LGSOC: low-grade serous ovarian cancer; MiT/TFE: microphthalmia family of transcription factors; NAC: N-acetylcysteine; PDAC: pancreatic ductal adenocarcinoma; ROS: reactive oxygen species; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joshua P. Gray
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiqun Mao
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hailing Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rumeysa Ozyurt
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivian R. Orellana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Cancer Research Institute Ghent, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Mark S. Carey
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
65
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
66
|
Ghosh C, Hu J. Importance of targeting various cell signaling pathways in solid cancers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:101-155. [PMID: 38663958 DOI: 10.1016/bs.ircmb.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Most adult human cancers are solid tumors prevailing in vital organs and lead to mortality all over the globe. Genetic and epigenetic alterations in cancer genes or genes of associated signaling pathways impart the most common characteristic of malignancy, that is, uncontrolled proliferation. Unless the mechanism of action of these cells signaling pathways (involved in cell proliferation, apoptosis, metastasis, and the maintenance of the stemness of cancer stem cells and cancer microenvironment) and their physiologic alteration are extensively studied, it is challenging to understand tumorigenesis as well as develop new treatments and precision medicines. Targeted therapy is one of the most promising strategies for treating various cancers. However, cancer is an evolving disease, and most patients develop resistance to these drugs by acquired mutations or mediation of microenvironmental factors or due to tumor heterogeneity. Researchers are striving to develop novel therapeutic options like combinatorial approaches targeting multiple responsible pathways effectively. Thus, in-depth knowledge of cell signaling and its components remains a critical topic of cancer research. This chapter summarized various extensively studied pathways in solid cancer and how they are targeted for therapeutic strategies.
Collapse
Affiliation(s)
- Chandrayee Ghosh
- Department of Surgery, Stanford University, Stanford, CA, Unites States.
| | - Jiangnan Hu
- Department of Surgery, Stanford University, Stanford, CA, Unites States
| |
Collapse
|
67
|
Scianò F, Terrana F, Pecoraro C, Parrino B, Cascioferro S, Diana P, Giovannetti E, Carbone D. Exploring the therapeutic potential of focal adhesion kinase inhibition in overcoming chemoresistance in pancreatic ductal adenocarcinoma. Future Med Chem 2024; 16:271-289. [PMID: 38269431 DOI: 10.4155/fmc-2023-0234] [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: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 01/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related deaths worldwide. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase often overexpressed in PDAC. FAK has been linked to cell migration, survival, proliferation, angiogenesis and adhesion. This review first highlights the chemoresistant nature of PDAC. Second, the role of FAK in PDAC cancer progression and resistance is carefully described. Additionally, it discusses recent developments of FAK inhibitors as valuable drugs in the treatment of PDAC, with a focus on diamine-substituted-2,4-pyrimidine-based compounds, which represent the most potent class of FAK inhibitors in clinical trials for the treatment of PDAC disease. To conclude, relevant computational studies performed on FAK inhibitors are reported to highlight the key structural features required for interaction with the protein, with the aim of optimizing this novel targeted therapy.
Collapse
Affiliation(s)
- Fabio Scianò
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Francesca Terrana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Camilla Pecoraro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Barbara Parrino
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Stella Cascioferro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Patrizia Diana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, San Giuliano Terme, Pisa, 56017, Italy
| | - Daniela Carbone
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| |
Collapse
|
68
|
Ureña E, Xu B, Regan JC, Atilano ML, Minkley LJ, Filer D, Lu YX, Bolukbasi E, Khericha M, Alic N, Partridge L. Trametinib ameliorates aging-associated gut pathology in Drosophila females by reducing Pol III activity in intestinal stem cells. Proc Natl Acad Sci U S A 2024; 121:e2311313121. [PMID: 38241436 PMCID: PMC10823232 DOI: 10.1073/pnas.2311313121] [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: 07/12/2023] [Accepted: 11/17/2023] [Indexed: 01/21/2024] Open
Abstract
Pharmacological therapies are promising interventions to slow down aging and reduce multimorbidity in the elderly. Studies in animal models are the first step toward translation of candidate molecules into human therapies, as they aim to elucidate the molecular pathways, cellular mechanisms, and tissue pathologies involved in the anti-aging effects. Trametinib, an allosteric inhibitor of MEK within the Ras/MAPK (Ras/Mitogen-Activated Protein Kinase) pathway and currently used as an anti-cancer treatment, emerged as a geroprotector candidate because it extended lifespan in the fruit fly Drosophila melanogaster. Here, we confirm that trametinib consistently and robustly extends female lifespan, and reduces intestinal stem cell (ISC) proliferation, tumor formation, tissue dysplasia, and barrier disruption in guts in aged flies. In contrast, pro-longevity effects of trametinib are weak and inconsistent in males, and it does not influence gut homeostasis. Inhibition of the Ras/MAPK pathway specifically in ISCs is sufficient to partially recapitulate the effects of trametinib. Moreover, in ISCs, trametinib decreases the activity of the RNA polymerase III (Pol III), a conserved enzyme synthesizing transfer RNAs and other short, non-coding RNAs, and whose inhibition also extends lifespan and reduces gut pathology. Finally, we show that the pro-longevity effect of trametinib in ISCs is partially mediated by Maf1, a repressor of Pol III, suggesting a life-limiting Ras/MAPK-Maf1-Pol III axis in these cells. The mechanism of action described in this work paves the way for further studies on the anti-aging effects of trametinib in mammals and shows its potential for clinical application in humans.
Collapse
Affiliation(s)
- Enric Ureña
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Bowen Xu
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Magda L. Atilano
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Lucy J. Minkley
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Danny Filer
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| | - Ekin Bolukbasi
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Mobina Khericha
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Nazif Alic
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| |
Collapse
|
69
|
Godfrey LK, Forster J, Liffers ST, Schröder C, Köster J, Henschel L, Ludwig KU, Lähnemann D, Trajkovic-Arsic M, Behrens D, Scarpa A, Lawlor RT, Witzke KE, Sitek B, Johnsen SA, Rahmann S, Horsthemke B, Zeschnigk M, Siveke JT. Pancreatic cancer acquires resistance to MAPK pathway inhibition by clonal expansion and adaptive DNA hypermethylation. Clin Epigenetics 2024; 16:13. [PMID: 38229153 PMCID: PMC10792938 DOI: 10.1186/s13148-024-01623-z] [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: 09/19/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis. It is marked by extraordinary resistance to conventional therapies including chemotherapy and radiation, as well as to essentially all targeted therapies evaluated so far. More than 90% of PDAC cases harbor an activating KRAS mutation. As the most common KRAS variants in PDAC remain undruggable so far, it seemed promising to inhibit a downstream target in the MAPK pathway such as MEK1/2, but up to now preclinical and clinical evaluation of MEK inhibitors (MEKi) failed due to inherent and acquired resistance mechanisms. To gain insights into molecular changes during the formation of resistance to oncogenic MAPK pathway inhibition, we utilized short-term passaged primary tumor cells from ten PDACs of genetically engineered mice. We followed gain and loss of resistance upon MEKi exposure and withdrawal by longitudinal integrative analysis of whole genome sequencing, whole genome bisulfite sequencing, RNA-sequencing and mass spectrometry data. RESULTS We found that resistant cell populations under increasing MEKi treatment evolved by the expansion of a single clone but were not a direct consequence of known resistance-conferring mutations. Rather, resistant cells showed adaptive DNA hypermethylation of 209 and hypomethylation of 8 genomic sites, most of which overlap with regulatory elements known to be active in murine PDAC cells. Both DNA methylation changes and MEKi resistance were transient and reversible upon drug withdrawal. Furthermore, MEKi resistance could be reversed by DNA methyltransferase inhibition with remarkable sensitivity exclusively in the resistant cells. CONCLUSION Overall, the concept of acquired therapy resistance as a result of the expansion of a single cell clone with epigenetic plasticity sheds light on genetic, epigenetic and phenotypic patterns during evolvement of treatment resistance in a tumor with high adaptive capabilities and provides potential for reversion through epigenetic targeting.
Collapse
Affiliation(s)
- Laura K Godfrey
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
| | - Jan Forster
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
- Genome Informatics, Institute of Human Genetics, University Duisburg-Essen, Essen, Germany
| | - Sven-Thorsten Liffers
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
| | - Christopher Schröder
- Genome Informatics, Institute of Human Genetics, University Duisburg-Essen, Essen, Germany
| | - Johannes Köster
- Bioinformatics and Computational Oncology, Institute for Artificial Intelligence in Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Leonie Henschel
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - David Lähnemann
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
| | - Marija Trajkovic-Arsic
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
| | - Diana Behrens
- EPO Experimental Pharmacology and Oncology GmbH, Berlin-Buch, Germany
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Pathological Anatomy Section, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Cancer Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Cancer Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Kathrin E Witzke
- Medizinisches Proteom-Center/Zentrum Für Protein-Diagnostik, Ruhr-Universität Bochum, Bochum, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center/Zentrum Für Protein-Diagnostik, Ruhr-Universität Bochum, Bochum, Germany
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Steven A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Sven Rahmann
- Algorithmic Bioinformatics, Center for Bioinformatics Saar and Saarland University, Saarland Informatics Campus, Saarbrücken, Germany
| | - Bernhard Horsthemke
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Michael Zeschnigk
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) West, Campus Essen, Essen, Germany.
| |
Collapse
|
70
|
Tripathi P, Kumari R, Pathak R. Drugging the undruggable: Advances in targeting KRAS signaling in solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:1-39. [PMID: 38663957 DOI: 10.1016/bs.ircmb.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Cancer remains the leading cause of global mortality, prompting a paradigm shift in its treatment and outcomes with the advent of targeted therapies. Among the most prevalent mutations in RAS-driven cancers, Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations account for approximately 86% of cases worldwide, particularly in lung, pancreatic, and colon cancers, contributing to poor prognosis and reduced overall survival. Despite numerous efforts to understand the biology of KRAS mutants and their pivotal role in cancer development, the lack of well-defined drug-binding pockets has deemed KRAS an "undruggable" therapeutic target, presenting significant challenges for researchers and clinicians alike. Through significant biochemical and technological advances, the last decade has witnessed promising breakthroughs in targeted therapies for KRAS-mutated lung, colon, and pancreatic cancers, marking a critical turning point in the field. In this chapter, we provide an overview of the characteristics of KRAS mutations across various solid tumors, highlighting ongoing cutting-edge research on the immune microenvironment, the development of KRAS-driven mice models, and the recent progress in the exploration of specific KRAS mutant-targeted therapeutic approaches. By comprehensive understanding of the intricacies of KRAS signaling in solid tumors and the latest therapeutic developments, this chapter will shed light on the potential for novel therapeutic strategies to combat KRAS-driven tumors and improve patient outcomes.
Collapse
Affiliation(s)
- Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, United States
| | - Rajni Kumari
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, United States.
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, United States.
| |
Collapse
|
71
|
Beilankouhi EAV, Valilo M, Dastmalchi N, Teimourian S, Safaralizadeh R. The Function of Autophagy in the Initiation, and Development of Breast Cancer. Curr Med Chem 2024; 31:2974-2990. [PMID: 37138421 DOI: 10.2174/0929867330666230503145319] [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: 12/09/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 05/05/2023]
Abstract
Autophagy is a significant catabolic procedure that increases in stressful conditions. This mechanism is mostly triggered after damage to the organelles, the presence of unnatural proteins, and nutrient recycling in reaction to these stresses. One of the key points in this article is that cleaning and preserving damaged organelles and accumulated molecules through autophagy in normal cells helps prevent cancer. Since dysfunction of autophagy is associated with various diseases, including cancer, it has a dual function in tumor suppression and expansion. It has newly become clear that the regulation of autophagy can be used for the treatment of breast cancer, which has a promising effect of increasing the efficiency of anticancer treatment in a tissue- and cell-type-specific manner by affecting the fundamental molecular mechanisms. Regulation of autophagy and its function in tumorigenesis is a vital part of modern anticancer techniques. This study discusses the current advances related to the mechanisms that describe essential modulators of autophagy involved in the metastasis of cancers and the development of new breast cancer treatments.
Collapse
Affiliation(s)
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
72
|
Kazi A, Ranjan A, Kumar M.V. V, Agianian B, Garcia Chavez M, Vudatha V, Wang R, Vangipurapu R, Chen L, Kennedy P, Subramanian K, Quirke JC, Beato F, Underwood PW, Fleming JB, Trevino J, Hergenrother PJ, Gavathiotis E, Sebti SM. Discovery of KRB-456, a KRAS G12D Switch-I/II Allosteric Pocket Binder That Inhibits the Growth of Pancreatic Cancer Patient-derived Tumors. CANCER RESEARCH COMMUNICATIONS 2023; 3:2623-2639. [PMID: 38051103 PMCID: PMC10754035 DOI: 10.1158/2767-9764.crc-23-0222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/26/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Currently, there are no clinically approved drugs that directly thwart mutant KRAS G12D, a major driver of human cancer. Here, we report on the discovery of a small molecule, KRB-456, that binds KRAS G12D and inhibits the growth of pancreatic cancer patient-derived tumors. Protein nuclear magnetic resonance studies revealed that KRB-456 binds the GDP-bound and GCP-bound conformation of KRAS G12D by forming interactions with a dynamic allosteric binding pocket within the switch-I/II region. Isothermal titration calorimetry demonstrated that KRB-456 binds potently to KRAS G12D with 1.5-, 2-, and 6-fold higher affinity than to KRAS G12V, KRAS wild-type, and KRAS G12C, respectively. KRB-456 potently inhibits the binding of KRAS G12D to the RAS-binding domain (RBD) of RAF1 as demonstrated by GST-RBD pulldown and AlphaScreen assays. Treatment of KRAS G12D-harboring human pancreatic cancer cells with KRB-456 suppresses the cellular levels of KRAS bound to GTP and inhibits the binding of KRAS to RAF1. Importantly, KRB-456 inhibits P-MEK, P-AKT, and P-S6 levels in vivo and inhibits the growth of subcutaneous and orthotopic xenografts derived from patients with pancreatic cancer whose tumors harbor KRAS G12D and KRAS G12V and who relapsed after chemotherapy and radiotherapy. These results warrant further development of KRB-456 for pancreatic cancer. SIGNIFICANCE There are no clinically approved drugs directly abrogating mutant KRAS G12D. Here, we discovered a small molecule, KRB-456, that binds a dynamic allosteric binding pocket within the switch-I/II region of KRAS G12D. KRB-456 inhibits P-MEK, P-AKT, and P-S6 levels in vivo and inhibits the growth of subcutaneous and orthotopic xenografts derived from patients with pancreatic cancer. This discovery warrants further advanced preclinical and clinical studies in pancreatic cancer.
Collapse
Affiliation(s)
- Aslamuzzaman Kazi
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida
| | - Alok Ranjan
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Vasantha Kumar M.V.
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Bogos Agianian
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Martin Garcia Chavez
- Department of Chemistry, Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Rui Wang
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | | | - Liwei Chen
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida
| | - Perry Kennedy
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida
| | - Karthikeyan Subramanian
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Jonathan C.K. Quirke
- Department of Chemistry, Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Francisca Beato
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | - Jason B. Fleming
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Jose Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Paul J. Hergenrother
- Department of Chemistry, Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Said M. Sebti
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
- Drug Discovery Department, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
73
|
Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther 2023; 8:455. [PMID: 38105263 PMCID: PMC10725898 DOI: 10.1038/s41392-023-01705-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 12/19/2023] Open
Abstract
Metastatic dissemination of solid tumors, a leading cause of cancer-related mortality, underscores the urgent need for enhanced insights into the molecular and cellular mechanisms underlying metastasis, chemoresistance, and the mechanistic backgrounds of individuals whose cancers are prone to migration. The most prevalent signaling cascade governed by multi-kinase inhibitors is the mitogen-activated protein kinase (MAPK) pathway, encompassing the RAS-RAF-MAPK kinase (MEK)-extracellular signal-related kinase (ERK) pathway. RAF kinase is a primary mediator of the MAPK pathway, responsible for the sequential activation of downstream targets, such as MEK and the transcription factor ERK, which control numerous cellular and physiological processes, including organism development, cell cycle control, cell proliferation and differentiation, cell survival, and death. Defects in this signaling cascade are associated with diseases such as cancer. RAF inhibitors (RAFi) combined with MEK blockers represent an FDA-approved therapeutic strategy for numerous RAF-mutant cancers, including melanoma, non-small cell lung carcinoma, and thyroid cancer. However, the development of therapy resistance by cancer cells remains an important barrier. Autophagy, an intracellular lysosome-dependent catabolic recycling process, plays a critical role in the development of RAFi resistance in cancer. Thus, targeting RAF and autophagy could be novel treatment strategies for RAF-mutant cancers. In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance. Furthermore, we explore and discuss the ongoing development of next-generation RAF inhibitors with enhanced therapeutic profiles. Additionally, this review sheds light on the functional interplay between RAF-targeted therapies and autophagy in cancer.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea.
| |
Collapse
|
74
|
Yi L, Luo M, Wang M, Dong Z, Du Y. Fangchinoline alleviates cognitive impairments through enhancing autophagy and mitigating oxidative stress in Alzheimer's disease models. Front Cell Dev Biol 2023; 11:1288506. [PMID: 38146492 PMCID: PMC10749363 DOI: 10.3389/fcell.2023.1288506] [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: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) is a debilitating, progressive, neurodegenerative disorder characterized by the deposition of amyloid-β (Aβ) peptides and subsequent oxidative stress, resulting in a cascade of cytotoxic effects. Fangchinoline (Fan), a bisbenzylisoquinoline alkaloid isolated from traditional Chinese herb Stephania tetrandra S. Moorec, has been reported to possess multiple potent biological activities, including anti-inflammatory and antioxidant properties. However, the potential neuroprotective efficacy of Fan against AD remains unknown. Methods: N2AAPP cells, the mouse neuroblastoma N2A cells stably transfected with human Swedish mutant APP695, were served as an in vitro AD model. A mouse model of AD was constructed by microinjection of Aβ1-42 peptides into lateral ventricle of WT mice. The neuroprotective effects of Fan on AD were investigated through a combination of Western blot analysis, immunoprecipitation and behavioral assessments. Results and discussion: It was found that Fan effectively attenuated the amyloidogenic processing of APP by augmenting autophagy and subsequently fostering lysosomal degradation of BACE1 in N2AAPP cells, as reflected by the decrease in P62 levels, concomitant with the increase in Beclin-1 and LC3-II levels. More importantly, Fan significantly ameliorated cognitive impairment in an Aβ1-42-induced mouse model of AD via the induction of autophagy and the inhibition of oxidative stress, as evidenced by an increase in antioxidants including glutathione reductase (GR), total antioxidant capacity (T-AOC), nuclear factor erythroid-2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and superoxide dismutase-1 (SOD-1) and a decrease in pro-oxidants including hydrogen peroxide (H2O2) and inducible nitric oxide synthase (i-NOS), coupled with a reduction in apoptosis marker, cleaved caspase-3. Taken together, our study demonstrate that Fan ameliorates cognitive dysfunction through promoting autophagy and mitigating oxidative stress, making it a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Lilin Yi
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Man Luo
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Maoju Wang
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Institute for Brain Science and Disease of Chongqing Medical University, Chongqing, China
| | - Yehong Du
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
75
|
Valdes Angues R, Perea Bustos Y. SARS-CoV-2 Vaccination and the Multi-Hit Hypothesis of Oncogenesis. Cureus 2023; 15:e50703. [PMID: 38234925 PMCID: PMC10792266 DOI: 10.7759/cureus.50703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex and dynamic disease. The "hallmarks of cancer" were proposed by Hanahan and Weinberg (2000) as a group of biological competencies that human cells attain as they progress from normalcy to neoplastic transformation. These competencies include self-sufficiency in proliferative signaling, insensitivity to growth-suppressive signals and immune surveillance, the ability to evade cell death, enabling replicative immortality, reprogramming energy metabolism, inducing angiogenesis, and activating tissue invasion and metastasis. Underlying these competencies are genome instability, which expedites their acquisition, and inflammation, which fosters their function(s). Additionally, cancer exhibits another dimension of complexity: a heterogeneous repertoire of infiltrating and resident host cells, secreted factors, and extracellular matrix, known as the tumor microenvironment, that through a dynamic and reciprocal relationship with cancer cells supports immortality, local invasion, and metastatic dissemination. This staggering intricacy calls for caution when advising all people with cancer (or a previous history of cancer) to receive the COVID-19 primary vaccine series plus additional booster doses. Moreover, because these patients were not included in the pivotal clinical trials, considerable uncertainty remains regarding vaccine efficacy, safety, and the risk of interactions with anticancer therapies, which could reduce the value and innocuity of either medical treatment. After reviewing the available literature, we are particularly concerned that certain COVID-19 vaccines may generate a pro-tumorigenic milieu (i.e., a specific environment that could lead to neoplastic transformation) that predisposes some (stable) oncologic patients and survivors to cancer progression, recurrence, and/or metastasis. This hypothesis is based on biological plausibility and fulfillment of the multi-hit hypothesis of oncogenesis (i.e., induction of lymphopenia and inflammation, downregulation of angiotensin-converting enzyme 2 (ACE2) expression, activation of oncogenic cascades, sequestration of tumor suppressor proteins, dysregulation of the RNA-G quadruplex-protein binding system, alteration of type I interferon responses, unsilencing of retrotransposable elements, etc.) together with growing evidence and safety reports filed to Vaccine Adverse Effects Report System (VAERS) suggesting that some cancer patients experienced disease exacerbation or recurrence following COVID-19 vaccination. In light of the above and because some of these concerns (i.e., alteration of oncogenic pathways, promotion of inflammatory cascades, and dysregulation of the renin-angiotensin system) also apply to cancer patients infected with SARS-CoV-2, we encourage the scientific and medical community to urgently evaluate the impact of both COVID-19 and COVID-19 vaccination on cancer biology and tumor registries, adjusting public health recommendations accordingly.
Collapse
Affiliation(s)
- Raquel Valdes Angues
- Neurology, Oregon Health and Science University School of Medicine, Portland, USA
| | | |
Collapse
|
76
|
Zhang P, Shi Y, Xu Y, Liang Y, Huang C, Zhong D, Zhang Z, Yu Y, Zhang Z, Zhang J, Yu L, Zuo Y, Wang X, Niu H. A Nano-Autophagy Inhibitor Triggering Reciprocal Feedback Control of Cholesterol Depletion for Solid Tumor Therapy. Adv Healthc Mater 2023; 12:e2302020. [PMID: 37767984 DOI: 10.1002/adhm.202302020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Solid tumors are characterized by enhanced metabolism of lipid, particularly cholesterol, inspiring the exploration of metabolic therapy through cholesterol oxidase (COD)-mediated cholesterol deprivation. However, the therapeutic efficacy of COD is limited due to the hypoxic tumor microenvironment and the protective autophagy triggered by cholesterol deprivation. Herein, a combination therapy for metabolically treating solid tumors through COD in conjunction with molybdenum oxide nanodots (MONDs), which serve as both potent oxygen generators and autophagy inhibitors, is reported. MONDs convert H2 O2 (arising from COD-mediated cholesterol oxidation) into O2 , which is then recycled by COD to form reciprocal feedback for cholesterol depletion. Concurrently, MONDs can overcome autophagy-induced therapeutic resistance frequently occurring in conventional nutrient deprivation therapy by activating AKT/mTOR pathway phosphorylation. Combination therapy in the xenograft model results in an ≈5-fold increase in therapeutic efficiency as compared with COD treatment alone. This functionally cooperative metabolic coupling strategy holds great promise as a novel polytherapy approach that will benefit patients with solid tumors.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yanfeng Shi
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Yuanhong Xu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Ye Liang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Chao Huang
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Di Zhong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Zhilei Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yongbo Yu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zhao Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jianfeng Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Lei Yu
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Yuhui Zuo
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Xinsheng Wang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Haitao Niu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| |
Collapse
|
77
|
Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. The role of mitochondrial dynamics in disease. MedComm (Beijing) 2023; 4:e462. [PMID: 38156294 PMCID: PMC10753647 DOI: 10.1002/mco2.462] [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: 09/18/2023] [Revised: 11/14/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Mitochondria are multifaceted and dynamic organelles regulating various important cellular processes from signal transduction to determining cell fate. As dynamic properties of mitochondria, fusion and fission accompanied with mitophagy, undergo constant changes in number and morphology to sustain mitochondrial homeostasis in response to cell context changes. Thus, the dysregulation of mitochondrial dynamics and mitophagy is unsurprisingly related with various diseases, but the unclear underlying mechanism hinders their clinical application. In this review, we summarize the recent developments in the molecular mechanism of mitochondrial dynamics and mitophagy, particularly the different roles of key components in mitochondrial dynamics in different context. We also summarize the roles of mitochondrial dynamics and target treatment in diseases related to the cardiovascular system, nervous system, respiratory system, and tumor cell metabolism demanding high-energy. In these diseases, it is common that excessive mitochondrial fission is dominant and accompanied by impaired fusion and mitophagy. But there have been many conflicting findings about them recently, which are specifically highlighted in this view. We look forward that these findings will help broaden our understanding of the roles of the mitochondrial dynamics in diseases and will be beneficial to the discovery of novel selective therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Xinyan Dai
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Hui Li
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huiling Jiang
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Junfu Zhou
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Shiying Zhang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jiacheng Guo
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Lidu Shen
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huantao Yang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jie Lin
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Hengxiu Yan
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| |
Collapse
|
78
|
Montenegro MF, Martí-Díaz R, Navarro A, Tolivia J, Sánchez-Del-Campo L, Cabezas-Herrera J, Rodríguez-López JN. Targeting protein methylation in pancreatic cancer cells results in KRAS signaling imbalance and inhibition of autophagy. Cell Death Dis 2023; 14:761. [PMID: 37996408 PMCID: PMC10667277 DOI: 10.1038/s41419-023-06288-9] [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: 07/04/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023]
Abstract
Pancreatic cancer cells with mutant KRAS require strong basal autophagy for viability and growth. Here, we observed that some processes that allow the maintenance of basal autophagy in pancreatic cancer cells are controlled by protein methylation. Thus, by maintaining the methylation status of proteins such as PP2A and MRAS, these cells can sustain their autophagic activity. Protein methylation disruption by a hypomethylating treatment (HMT), which depletes cellular S-adenosylmethionine levels while inducing S-adenosylhomocysteine accumulation, resulted in autophagy inhibition and endoplasmic reticulum stress-induced apoptosis in pancreatic cancer cells. We observed that by reducing the membrane localization of MRAS, hypomethylation conditions produced an imbalance in KRAS signaling, resulting in the partial inactivation of ERK and hyperactivation of the PI3K/AKT-mTORC1 pathway. Interestingly, HMT impeded CRAF activation by disrupting the ternary SHOC2 complex (SHOC2/MRAS/PP1), which functions as a CRAF-S259 holophosphatase. The demethylation events that resulted in PP2A inactivation also favored autophagy inhibition by preventing ULK1 activation while restoring the cytoplasmic retention of the MiT/TFE transcription factors. Since autophagy provides pancreatic cancer cells with metabolic plasticity to cope with various metabolic stress conditions, while at the same time promoting their pathogenesis and resistance to KRAS pathway inhibitors, this hypomethylating treatment could represent a therapeutic opportunity for pancreatic adenocarcinomas.
Collapse
Affiliation(s)
- María F Montenegro
- Department of Biochemistry and Molecular Biology A, School of Biology, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain.
| | - Román Martí-Díaz
- Department of Biochemistry and Molecular Biology A, School of Biology, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Ana Navarro
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Grupo GECYEN del Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Jorge Tolivia
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Grupo GECYEN del Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Luis Sánchez-Del-Campo
- Department of Biochemistry and Molecular Biology A, School of Biology, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Juan Cabezas-Herrera
- Molecular Therapy and Biomarkers Research Group, University Hospital Virgen de la Arrixaca, IMIB, Murcia, Spain
| | - José Neptuno Rodríguez-López
- Department of Biochemistry and Molecular Biology A, School of Biology, University of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain.
| |
Collapse
|
79
|
Huang S, Lin L, Ma Y, Zhu Q, Weng N. Scoparone induces autophagic cell death via the PAK1/AKT axis in colorectal cancer. Eur J Pharmacol 2023; 959:176091. [PMID: 37805132 DOI: 10.1016/j.ejphar.2023.176091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
Colorectal cancer (CRC) is one of most common malignancies worldwide, yet curative therapy remains a clinical challenge. Here, we demonstrate that scoparone (Scop), a traditional Chinese medicine monomer, inhibits the growth of CRC cells both in vitro and in vivo. Further studies found that Scop treatment induces complete autophagic flux in CRC cells, while inhibition of autophagy markedly represses the antiproliferative activities of Scop, suggesting an antitumour property of Scop-induced autophagy in CRC. Mechanistically, Scop induced autophagy initiation by reducing P21-activated kinase 1 (PAK1) expression and subsequently repressing the AKT/mTOR signaling pathway. Collectively, our study suggests that Scop is a potential anti-CRC therapeutic option and provides an underlying molecular mechanism for its antitumour effect in CRC.
Collapse
Affiliation(s)
- Sha Huang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China
| | - Luping Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China
| | - Yifei Ma
- Department of Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Ningna Weng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, 350011, PR China; Department of Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
80
|
Huang Y, Zhen Y, Chen Y, Sui S, Zhang L. Unraveling the interplay between RAS/RAF/MEK/ERK signaling pathway and autophagy in cancer: From molecular mechanisms to targeted therapy. Biochem Pharmacol 2023; 217:115842. [PMID: 37802240 DOI: 10.1016/j.bcp.2023.115842] [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: 07/24/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
RAS/RAF/MEK/ERK signaling pathway is one of the most important pathways of Mitogen-activated protein kinases (MAPK), which widely participate in regulating cell proliferation, differentiation, apoptosis and signaling transduction. Autophagy is an essential mechanism that maintains cellular homeostasis by degrading aged and damaged organelles. Recently, some studies revealed RAS/RAF/MEK/ERK signaling pathway is closely related to autophagy regulation and has a dual effect in tumor cells. However, the specific mechanism by which RAS/RAF/MEK/ERK signaling pathway participates in autophagy regulation is not fully understood. This article provides a comprehensive review of the research progress with regard to the RAS/RAF/MEK/ERK signaling pathway and autophagy, as well as their interplay in cancer therapy. The impact of small molecule inhibitors that target the RAS/RAF/MEK/ERK signaling pathway on autophagy is discussed in this study. The advantages and limitations of the clinical combination of these small molecule inhibitors with autophagy inhibitors are also explored. The findings from this study may provide additional perspectives for future cancer treatment strategies.
Collapse
Affiliation(s)
- Yunli Huang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yongqi Zhen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanmei Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shaoguang Sui
- Emergency Department, The Second Hospital, Dalian Medical University, Dalian 116000, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
81
|
Wang H, Liang Y, Zhang T, Yu X, Song X, Chen Y, Mao Q, Xia W, Chen B, Xu L, Dong G, Jiang F. C-IGF1R encoded by cIGF1R acts as a molecular switch to restrict mitophagy of drug-tolerant persister tumour cells in non-small cell lung cancer. Cell Death Differ 2023; 30:2365-2381. [PMID: 37689814 PMCID: PMC10657401 DOI: 10.1038/s41418-023-01222-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/11/2023] Open
Abstract
The clinical efficacy of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors (EGFR-TKIs) is limited by the emergence of drug resistance. We hypothesise that restoring dysregulated circular RNAs under initial treatment with EGFR-TKIs may enhance their effectiveness. Through high-throughput screening, we identify that combining circular RNA IGF1R (cIGF1R) with EGFR-TKIs significantly synergises to suppress tumour regrowth following drug withdrawal. Mechanistically, cIGF1R interacts with RNA helicase A (RHA) to depress insulin-like growth factor 1 receptor (IGF1R) mRNA splicing, negatively regulating the parent IGF1R signalling pathway. This regulation is similar to that of IGF1R inhibitor, which induces drug-tolerant persister (DTP) state with activated mitophagy. The cIGF1R also encodes a peptide C-IGF1R that reduces Parkin-mediated ubiquitination of voltage-dependent anion channel 1 (VDAC1) to restrict mitophagy, acting as a molecular switch that promotes the transition of DTP to apoptosis. Our study shows that combining cIGF1R with EGFR-TKIs efficiently reduces the emergence of DTP.
Collapse
Affiliation(s)
- Hui Wang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yingkuan Liang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Te Zhang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Collaborative Innovation Centre for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Xinnian Yu
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
| | - Xuming Song
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Yuzhong Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Qixing Mao
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
| | - Wenjie Xia
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
| | - Bing Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
| | - Lin Xu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Gaochao Dong
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China.
| | - Feng Jiang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China.
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing, 210009, China.
| |
Collapse
|
82
|
Yamamoto K, Iwadate D, Naito E, Tateishi K, Fujishiro M. Autophagy as a critical driver of metabolic adaptation, therapeutic resistance, and immune evasion of cancer. Curr Opin Biotechnol 2023; 84:103012. [PMID: 39492353 DOI: 10.1016/j.copbio.2023.103012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 11/05/2024]
Abstract
Autophagy is a well-conserved intracellular degradation pathway. Besides its physiological role in normal cells, autophagy is activated in various cancer types and protects cancer cells from stresses such as nutrient deprivation, therapeutic insults, and antitumor immunity. Autophagy in cancer cells as well as normal cells in the host supports tumor metabolism, allowing for tumor growth under a nutrient-limited tumor microenvironment. Autophagy also protects cancer cells from treatments such as radiation therapy, cytotoxic chemotherapy, and targeted therapy. Though the roles of autophagy in antitumor immunity are complex and highly context-dependent, accumulating evidence now supports the role of autophagy in mediating immunotherapy resistance. Based on these preclinical findings, multiple clinical trials are currently ongoing to test the therapeutic efficacy of autophagy inhibition in cancer. Here, we review recent findings on the tumor-promoting roles of autophagy in cancer and discuss advances in therapeutic approaches that target autophagy in cancer.
Collapse
Affiliation(s)
- Keisuke Yamamoto
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Dosuke Iwadate
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Eri Naito
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, St. Marianna University School of Medicine, 2-16-1 Sugao, Kawasaki city, Kanagawa 216-8511 Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
83
|
Meng F, Song C, Liu J, Chen F, Zhu Y, Fang X, Cao Q, Ma D, Wang Y, Zhang C. Chlorogenic Acid Modulates Autophagy by Inhibiting the Activity of ALKBH5 Demethylase, Thereby Ameliorating Hepatic Steatosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15073-15086. [PMID: 37805933 DOI: 10.1021/acs.jafc.3c03710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Chlorogenic acid (CGA) is a naturally occurring plant component with the purpose of alleviating hepatic lipid deposition biological activities. However, the molecular mechanism behind this ability of CGA remains unelucidated. Consequently, we investigated the effect of CGA on hepatic lipid accumulation and elucidated its underlying mechanism. Our study used a high-fat diet (HFD)-induced mouse nonalcoholic fatty liver disease (NAFLD) model in mice to investigate the impact of CGA on hepatic lipid accumulation. The results revealed that the oral administration of CGA can ameliorate HFD-induced hepatic lipid deposition, reduce the NAFLD activity score (NAS), enhance liver autophagy, mitigate liver cell structural damage, and inhibit the MAPK/ERK signaling pathway. Meanwhile, CGA treatment increased the LC3B:LC3B ratio and decreased P62 expression. Cell experiments demonstrated that autophagy contributes to the ability of CGA to alleviate lipid deposition. Further analysis revealed that CGA specifically binds to ALKBH5 and inhibits its m6A methylase activity. The inhibition of ALKBH5 activity significantly reduces AXL mRNA stability in liver cells. The AXL downregulation resulted in suppressing ERK signaling pathway activation. Overall, this study demonstrates that CGA can alleviate hepatic steatosis by regulating autophagy through the inhibition of ALKBH5 activity inhibition.
Collapse
Affiliation(s)
- Fantong Meng
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - Chengchuang Song
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - Jia Liu
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - Fang Chen
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - YuHua Zhu
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Xingtang Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - Qinghe Cao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
- Sweetpotato Research Institute, Chinese Academy of Agricultural Sciences, Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai District, Xuzhou, Jiangsu Province 221004, China
| | - Daifu Ma
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
- Sweetpotato Research Institute, Chinese Academy of Agricultural Sciences, Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai District, Xuzhou, Jiangsu Province 221004, China
| | - Yanhong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| | - Chunlei Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Institute of Cellular and Molecular Biology, College of Life Science, Jiangsu Normal University, Xuzhou Jiangsu Province, 221116, China
| |
Collapse
|
84
|
de Jesus VHF, Mathias-Machado MC, de Farias JPF, Aruquipa MPS, Jácome AA, Peixoto RD. Targeting KRAS in Pancreatic Ductal Adenocarcinoma: The Long Road to Cure. Cancers (Basel) 2023; 15:5015. [PMID: 37894382 PMCID: PMC10605759 DOI: 10.3390/cancers15205015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains an important cause of cancer-related mortality, and it is expected to play an even bigger part in cancer burden in the years to come. Despite concerted efforts from scientists and physicians, patients have experienced little improvement in survival over the past decades, possibly because of the non-specific nature of the tested treatment modalities. Recently, the discovery of potentially targetable molecular alterations has paved the way for the personalized treatment of PDAC. Indeed, the central piece in the molecular framework of PDAC is starting to be unveiled. KRAS mutations are seen in 90% of PDACs, and multiple studies have demonstrated their pivotal role in pancreatic carcinogenesis. Recent investigations have shed light on the differences in prognosis as well as therapeutic implications of the different KRAS mutations and disentangled the relationship between KRAS and effectors of downstream and parallel signaling pathways. Additionally, the recognition of other mechanisms involving KRAS-mediated pathogenesis, such as KRAS dosing and allelic imbalance, has contributed to broadening the current knowledge regarding this molecular alteration. Finally, KRAS G12C inhibitors have been recently tested in patients with pancreatic cancer with relative success, and inhibitors of KRAS harboring other mutations are under clinical development. These drugs currently represent a true hope for a meaningful leap forward in this dreadful disease.
Collapse
Affiliation(s)
| | | | | | | | - Alexandre A. Jácome
- Department of Gastrointestinal Medical Oncology, Oncoclínicas, Belo Horizonte 30360-680, Brazil
| | | |
Collapse
|
85
|
Koh Y, Kim H, Joo SY, Song S, Choi YH, Kim HR, Moon B, Byun J, Hong J, Shin DY, Park S, Lee KH, Lee KT, Lee JK, Park D, Lee SH, Jang JY, Lee H, Kim JA, Yoon SS, Park JK. Genetic assessment of pathogenic germline alterations in lysosomal genes among Asian patients with pancreatic ductal adenocarcinoma. J Transl Med 2023; 21:730. [PMID: 37848935 PMCID: PMC10580633 DOI: 10.1186/s12967-023-04549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Lysosomes are closely linked to autophagic activity, which plays a vital role in pancreatic ductal adenocarcinoma (PDAC) biology. The survival of PDAC patients is still poor, and the identification of novel genetic factors for prognosis and treatment is highly required to prevent PDAC-related deaths. This study investigated the germline variants related to lysosomal dysfunction in patients with PDAC and to analyze whether they contribute to the development of PDAC. METHODS The germline putative pathogenic variants (PPV) in genes involved in lysosomal storage disease (LSD) was compared between patients with PDAC (n = 418) and healthy controls (n = 845) using targeted panel and whole-exome sequencing. Furthermore, pancreatic organoids from wild-type and KrasG12D mice were used to evaluate the effect of lysosomal dysfunction on PDAC development. RNA sequencing (RNA-seq) analysis was performed with established PDAC patient-derived organoids (PDOs) according to the PPV status. RESULTS The PPV in LSD-related genes was higher in patients with PDAC than in healthy controls (8.13 vs. 4.26%, Log2 OR = 1.65, P = 3.08 × 10-3). The PPV carriers of LSD-related genes with PDAC were significantly younger than the non-carriers (mean age 61.5 vs. 65.3 years, P = 0.031). We further studied a variant of the lysosomal enzyme, galactosylceramidase (GALC), which was the most frequently detected LSD variant in our cohort. Autophagolysosomal activity was hampered when GALC was downregulated, which was accompanied by paradoxically elevated autophagic flux. Furthermore, the number of proliferating Ki-67+ cells increased significantly in pancreatic organoids derived from Galc knockout KrasG12D mice. Moreover, GALC PPV carriers tended to show drug resistance in both PDAC cell line and PDAC PDO, and RNA-seq analysis revealed that various metabolism and gene repair pathways were upregulated in PDAC PDOs harboring a GALC variant. CONCLUSIONS Genetically defined lysosomal dysfunction is frequently observed in patients with young-onset PDAC. This might contribute to PDAC development by altering metabolism and impairing autophagolysosomal activity, which could be potentially implicated in therapeutic applications for PDAC.
Collapse
Affiliation(s)
- Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyemin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - So Young Joo
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Seulki Song
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young Hoon Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Rae Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byul Moon
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jamin Byun
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Solip Park
- Structural Biology Department, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Kwang Hyuck Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyu Taek Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Kyun Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Daechan Park
- Department of Molecular Science and Technology, Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Se-Hoon Lee
- Department of Hematology/Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Departments of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Hyunsook Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea.
| | - Jung-Ae Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea.
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University School of Medicine, Seoul, Republic of Korea.
| | - Joo Kyung Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
86
|
Tang H, Ge Y, You T, Li X, Wang Y, Cheng Y, Bai C. A real-world analysis of trametinib in combination with hydroxychloroquine or CDK4/6 inhibitor as third- or later-line therapy in metastatic pancreatic adenocarcinoma. BMC Cancer 2023; 23:958. [PMID: 37817078 PMCID: PMC10563303 DOI: 10.1186/s12885-023-11464-3] [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: 05/11/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND There are no standard third-line treatment options for metastatic pancreatic ductal adenocarcinoma (mPDAC). Trametinib in combination with hydroxychloroquine (HCQ) or CDK4/6 inhibitors for pancreatic adenocarcinoma showed promising efficacy in preclinical studies. However, the regimens have not been well examined in patients with mPDAC. METHODS Patients with mPDAC who received the combination of trametinib and HCQ or CDK4/6 inhibitors as third- or later-line therapy were reviewed. The efficacy and prognosis were further analyzed. RESULTS A total of 13 mPDAC patients were enrolled, of whom 8 and 5 patients were treated with trametinib plus HCQ or a CDK4/6 inhibitor (palbociclib or abemaciclib), respectively. All enrolled patients had either KRAS G12D or G12V mutations and had received a median of 3 prior lines of therapy (range, 2-6). The median trametinib treatment duration was 1.4 months. Of the 10 patients with measurable disease, only 1 patient achieved stable disease, and the remaining patients had progressive disease. Moreover, in patients treated with trametinib plus HCQ and a CDK4/6 inhibitor, the median progression-free survival was 2.0 and 2.8 months, respectively, and the median overall survival was 4.2 and 4.7 months, respectively. Moreover, 5 (50%) patients experienced grade 3-4 adverse events in 10 patients with available safety data. CONCLUSIONS The combination of trametinib and HCQ or CDK4/6 inhibitors may not be an effective later-line treatment for mPDAC, and the current preliminary findings need to be confirmed by other studies with larger sample sizes.
Collapse
Affiliation(s)
- Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoyuan Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
87
|
Macaya I, Roman M, Welch C, Entrialgo-Cadierno R, Salmon M, Santos A, Feliu I, Kovalski J, Lopez I, Rodriguez-Remirez M, Palomino-Echeverria S, Lonfgren SM, Ferrero M, Calabuig S, Ludwig IA, Lara-Astiaso D, Jantus-Lewintre E, Guruceaga E, Narayanan S, Ponz-Sarvise M, Pineda-Lucena A, Lecanda F, Ruggero D, Khatri P, Santamaria E, Fernandez-Irigoyen J, Ferrer I, Paz-Ares L, Drosten M, Barbacid M, Gil-Bazo I, Vicent S. Signature-driven repurposing of Midostaurin for combination with MEK1/2 and KRASG12C inhibitors in lung cancer. Nat Commun 2023; 14:6332. [PMID: 37816716 PMCID: PMC10564741 DOI: 10.1038/s41467-023-41828-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Drug combinations are key to circumvent resistance mechanisms compromising response to single anti-cancer targeted therapies. The implementation of combinatorial approaches involving MEK1/2 or KRASG12C inhibitors in the context of KRAS-mutated lung cancers focuses fundamentally on targeting KRAS proximal activators or effectors. However, the antitumor effect is highly determined by compensatory mechanisms arising in defined cell types or tumor subgroups. A potential strategy to find drug combinations targeting a larger fraction of KRAS-mutated lung cancers may capitalize on the common, distal gene expression output elicited by oncogenic KRAS. By integrating a signature-driven drug repurposing approach with a pairwise pharmacological screen, here we show synergistic drug combinations consisting of multi-tyrosine kinase PKC inhibitors together with MEK1/2 or KRASG12C inhibitors. Such combinations elicit a cytotoxic response in both in vitro and in vivo models, which in part involves inhibition of the PKC inhibitor target AURKB. Proteome profiling links dysregulation of MYC expression to the effect of both PKC inhibitor-based drug combinations. Furthermore, MYC overexpression appears as a resistance mechanism to MEK1/2 and KRASG12C inhibitors. Our study provides a rational framework for selecting drugs entering combinatorial strategies and unveils MEK1/2- and KRASG12C-based therapies for lung cancer.
Collapse
Affiliation(s)
- Irati Macaya
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Marta Roman
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Connor Welch
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | | | - Marina Salmon
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Alba Santos
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Iker Feliu
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Joanna Kovalski
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Ines Lopez
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Maria Rodriguez-Remirez
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Sara Palomino-Echeverria
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra, Pamplona, Spain
| | - Shane M Lonfgren
- Stanford Institute for Immunity, Transplantation and Infection, Stanford, CA, USA
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Macarena Ferrero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
| | - Silvia Calabuig
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
- Department of Pathology, Universitat de Valencia, Valencia, Spain
| | - Iziar A Ludwig
- University of Navarra, Center for Applied Medical Research, Molecular Therapies Program, Pamplona, Spain
| | - David Lara-Astiaso
- University of Navarra, Center for Applied Medical Research, Genomics Platform, Pamplona, Spain
| | - Eloisa Jantus-Lewintre
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
- Department of Pathology, Universitat de Valencia, Valencia, Spain
| | - Elizabeth Guruceaga
- University of Navarra, Center for Applied Medical Research, Bioinformatics Platform, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Shruthi Narayanan
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
| | - Antonio Pineda-Lucena
- University of Navarra, Center for Applied Medical Research, Molecular Therapies Program, Pamplona, Spain
| | - Fernando Lecanda
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Purvesh Khatri
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra, Pamplona, Spain
| | - Enrique Santamaria
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joaquin Fernandez-Irigoyen
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Irene Ferrer
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Paz-Ares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Medical School, Universidad Complutense, Madrid, Spain
| | - Matthias Drosten
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain
| | - Mariano Barbacid
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Gil-Bazo
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
- Department of Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Silve Vicent
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
- University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain.
| |
Collapse
|
88
|
Yuan W, Fang W, Zhang R, Lyu H, Xiao S, Guo D, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Therapeutic strategies targeting AMPK-dependent autophagy in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119537. [PMID: 37463638 DOI: 10.1016/j.bbamcr.2023.119537] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
Macroautophagy is a health-modifying process of engulfing misfolded or aggregated proteins or damaged organelles, coating these proteins or organelles into vesicles, fusion of vesicles with lysosomes to form autophagic lysosomes, and degradation of the encapsulated contents. It is also a self-rescue strategy in response to harsh environments and plays an essential role in cancer cells. AMP-activated protein kinase (AMPK) is the central pathway that regulates autophagy initiation and autophagosome formation by phosphorylating targets such as mTORC1 and unc-51 like activating kinase 1 (ULK1). AMPK is an evolutionarily conserved serine/threonine protein kinase that acts as an energy sensor in cells and regulates various metabolic processes, including those involved in cancer. The regulatory network of AMPK is complicated and can be regulated by multiple upstream factors, such as LKB1, AKT, PPAR, SIRT1, or noncoding RNAs. Currently, AMPK is being investigated as a novel target for anticancer therapies based on its role in macroautophagy regulation. Herein, we review the effects of AMPK-dependent autophagy on tumor cell survival and treatment strategies targeting AMPK.
Collapse
Affiliation(s)
- Wenbin Yuan
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Wanyi Fang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rui Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dong Guo
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| |
Collapse
|
89
|
Sathiyaseelan P, Chittaranjan S, Kalloger SE, Chan J, Go NE, Jardon MA, Ho CJ, Hui T, Xu J, Chow C, Gao D, Johnson FD, Lockwood WW, Morin GB, Renouf DJ, Schaeffer DF, Gorski SM. Loss of ATG4B and ATG4A results in two-stage cell cycle defects in pancreatic ductal adenocarcinoma cells. J Cell Sci 2023; 136:jcs260644. [PMID: 37701987 PMCID: PMC10617609 DOI: 10.1242/jcs.260644] [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: 09/23/2022] [Accepted: 07/17/2023] [Indexed: 09/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits elevated levels of autophagy, which promote tumor progression and treatment resistance. ATG4B is an autophagy-related cysteine protease under consideration as a potential therapeutic target, but it is largely unexplored in PDAC. Here, we investigated the clinical and functional relevance of ATG4B expression in PDAC. Using two PDAC patient cohorts, we found that low ATG4B mRNA or protein expression is associated with worse patient survival outcomes, poorly differentiated PDAC tumors and a lack of survival benefit from adjuvant chemotherapy. In PDAC cell lines, ATG4B knockout reduced proliferation, abolished processing of LC3B (also known as MAP1LC3B), and reduced GABARAP and GABARAPL1 levels, but increased ATG4A levels. ATG4B and ATG4A double knockout lines displayed a further reduction in proliferation, characterized by delays in G1-S phase transition and mitosis. Pro-LC3B accumulated aberrantly at the centrosome with a concomitant increase in centrosomal proteins PCM1 and CEP131, which was rescued by exogenous ATG4B. The two-stage cell cycle defects following ATG4B and ATG4A loss have important therapeutic implications for PDAC.
Collapse
Affiliation(s)
- Paalini Sathiyaseelan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Suganthi Chittaranjan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Steve E. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
| | - Jennifer Chan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Nancy E. Go
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Mario A. Jardon
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Cally J. Ho
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Theodore Hui
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Jing Xu
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver, BC, V6H 3Z6, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, Vancouver, BC, V6H 3Z6, Canada
| | - Fraser D. Johnson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Integrative Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - William W. Lockwood
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- Department of Integrative Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Gregg B. Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Daniel J. Renouf
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
- Division of Medical Oncology, BC Cancer, Vancouver, BC, V5Z 4E6, Canada
| | - David F. Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
- Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada
| | - Sharon M. Gorski
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 3N1, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| |
Collapse
|
90
|
Barnaba C, Broadbent DG, Perez GI, Schmidt JC. AMPK Regulates Phagophore-to-Autophagosome Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559981. [PMID: 37808644 PMCID: PMC10557706 DOI: 10.1101/2023.09.28.559981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Autophagy is an important metabolic pathway that can non-selectively recycle cellular material or lead to targeted degradation of protein aggregates or damaged organelles. Autophagosome formation starts with autophagy factors accumulating on lipid vesicles containing ATG9. These phagophores attach to donor membranes, expand via ATG2-mediated lipid transfer, capture cargo, and mature into autophagosomes, ultimately fusing with lysosomes for their degradation. Autophagy can be activated by nutrient stress, for example by a reduction in the cellular levels of amino acids. In contrast, how autophagy is regulated by low cellular ATP levels via the AMP-activated protein kinase (AMPK), an important therapeutic target, is less clear. Using live-cell imaging and an automated image analysis pipeline, we systematically dissect how nutrient starvation regulates autophagosome biogenesis. We demonstrate that glucose starvation downregulates autophagosome maturation by AMPK mediated inhibition of phagophores tethering to donor membranes. Our results clarify AMPK's regulatory role in autophagy and highlight its potential as a therapeutic target to reduce autophagy.
Collapse
Affiliation(s)
- Carlo Barnaba
- Institute for Quantitative Health Science and Engineering
| | - David G. Broadbent
- Institute for Quantitative Health Science and Engineering
- College of Osteopathic Medicine
- Department of Physiology
| | | | - Jens C. Schmidt
- Institute for Quantitative Health Science and Engineering
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, USA
| |
Collapse
|
91
|
Taraborrelli L, Şenbabaoğlu Y, Wang L, Lim J, Blake K, Kljavin N, Gierke S, Scherl A, Ziai J, McNamara E, Owyong M, Rao S, Calviello AK, Oreper D, Jhunjhunwala S, Argiles G, Bendell J, Kim TW, Ciardiello F, Wongchenko MJ, de Sauvage FJ, de Sousa E Melo F, Yan Y, West NR, Murthy A. Tumor-intrinsic expression of the autophagy gene Atg16l1 suppresses anti-tumor immunity in colorectal cancer. Nat Commun 2023; 14:5945. [PMID: 37741832 PMCID: PMC10517947 DOI: 10.1038/s41467-023-41618-7] [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: 01/31/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023] Open
Abstract
Microsatellite-stable colorectal cancer (MSS-CRC) is highly refractory to immunotherapy. Understanding tumor-intrinsic determinants of immunotherapy resistance is critical to improve MSS-CRC patient outcomes. Here, we demonstrate that high tumor expression of the core autophagy gene ATG16L1 is associated with poor clinical response to anti-PD-L1 therapy in KRAS-mutant tumors from IMblaze370 (NCT02788279), a large phase III clinical trial of atezolizumab (anti-PD-L1) in advanced metastatic MSS-CRC. Deletion of Atg16l1 in engineered murine colon cancer organoids inhibits tumor growth in primary (colon) and metastatic (liver and lung) niches in syngeneic female hosts, primarily due to increased sensitivity to IFN-γ-mediated immune pressure. ATG16L1 deficiency enhances programmed cell death of colon cancer organoids induced by IFN-γ and TNF, thus increasing their sensitivity to host immunity. In parallel, ATG16L1 deficiency reduces tumor stem-like populations in vivo independently of adaptive immune pressure. This work reveals autophagy as a clinically relevant mechanism of immune evasion and tumor fitness in MSS-CRC and provides a rationale for autophagy inhibition to boost immunotherapy responses in the clinic.
Collapse
Affiliation(s)
- Lucia Taraborrelli
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA
| | - Yasin Şenbabaoğlu
- Department of Oncology Bioinformatics, Genentech Inc., South San Francisco, USA
| | - Lifen Wang
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA
| | - Junghyun Lim
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA
| | - Kerrigan Blake
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA
| | - Noelyn Kljavin
- Department of Molecular Oncology, Genentech Inc., South San Francisco, USA
| | - Sarah Gierke
- Center for Advanced Light Microscopy, Genentech Inc., South San Francisco, USA
- Department of Pathology, Genentech Inc., South San Francisco, USA
| | - Alexis Scherl
- Department of Pathology, Genentech Inc., South San Francisco, USA
| | - James Ziai
- Department of Pathology, Genentech Inc., South San Francisco, USA
| | - Erin McNamara
- Department of In Vivo Pharmacology, Genentech Inc., South San Francisco, USA
| | - Mark Owyong
- Department of In Vivo Pharmacology, Genentech Inc., South San Francisco, USA
| | - Shilpa Rao
- Department of Oncology Bioinformatics, Genentech Inc., South San Francisco, USA
| | | | - Daniel Oreper
- Department of Oncology Bioinformatics, Genentech Inc., South San Francisco, USA
| | - Suchit Jhunjhunwala
- Department of Oncology Bioinformatics, Genentech Inc., South San Francisco, USA
| | - Guillem Argiles
- Vall d'Hebrón Institute of Oncology, Vall d'Hebrón University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - Tae Won Kim
- Department of Oncology, Medical Center, University of Ulsan, Seoul, Korea
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | | | | | | | - Yibing Yan
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Nathaniel R West
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA.
| | - Aditya Murthy
- Department of Cancer Immunology, Genentech Inc., South San Francisco, USA.
- Gilead Sciences, Foster City, USA.
| |
Collapse
|
92
|
Lee E, Archasappawat S, Ji K, Pena J, Fernandez-Vega V, Gangaraju R, Beesabathuni NS, Kim MJ, Tian Q, Shah PS, Scampavia L, Spicer TP, Hwang CI. A new vulnerability to BET inhibition due to enhanced autophagy in BRCA2 deficient pancreatic cancer. Cell Death Dis 2023; 14:620. [PMID: 37735513 PMCID: PMC10514057 DOI: 10.1038/s41419-023-06145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Pancreatic cancer is one of the deadliest diseases in human malignancies. Among total pancreatic cancer patients, ~10% of patients are categorized as familial pancreatic cancer (FPC) patients, carrying germline mutations of the genes involved in DNA repair pathways (e.g., BRCA2). Personalized medicine approaches tailored toward patients' mutations would improve patients' outcome. To identify novel vulnerabilities of BRCA2-deficient pancreatic cancer, we generated isogenic Brca2-deficient murine pancreatic cancer cell lines and performed high-throughput drug screens. High-throughput drug screening revealed that Brca2-deficient cells are sensitive to Bromodomain and Extraterminal Motif (BET) inhibitors, suggesting that BET inhibition might be a potential therapeutic approach. We found that BRCA2 deficiency increased autophagic flux, which was further enhanced by BET inhibition in Brca2-deficient pancreatic cancer cells, resulting in autophagy-dependent cell death. Our data suggests that BET inhibition can be a novel therapeutic strategy for BRCA2-deficient pancreatic cancer.
Collapse
Affiliation(s)
- EunJung Lee
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, Davis, CA, 95616, USA
| | - Keely Ji
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Jocelyn Pena
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Virneliz Fernandez-Vega
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Ritika Gangaraju
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Nitin Sai Beesabathuni
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Martin Jean Kim
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Qi Tian
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Priya S Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Louis Scampavia
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Timothy P Spicer
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
93
|
Beatty GL, Delman D, Yu J, Liu M, Li JH, Zhang L, Lee JW, Chang RB, Bahary N, Kennedy EP, Wang-Gillam A, Rossi GR, Garrido-Laguna I. Treatment Response in First-Line Metastatic Pancreatic Ductal Adenocarcinoma Is Stratified By a Composite Index of Tumor Proliferation and CD8 T-Cell Infiltration. Clin Cancer Res 2023; 29:3514-3525. [PMID: 37534996 PMCID: PMC10530235 DOI: 10.1158/1078-0432.ccr-23-0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/16/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
PURPOSE Determinants of treatment outcomes to chemotherapy-based regimens in metastatic pancreatic ductal adenocarcinoma (PDA) remain ill-defined. Our aim was to examine tissue-based correlates of treatment response and resistance using matched baseline and on-treatment biopsies collected from patients with PDA treated in the first-line metastatic setting. EXPERIMENTAL DESIGN Patients with treatment-naïve metastatic PDA were enrolled in a Phase II trial (NCT02077881) investigating gemcitabine plus nab-paclitaxel in combination with indoximod, an orally administered small-molecule inhibitor of the IDO pathway. Baseline and on-treatment biopsies (week 8) of metastatic lesions (88% liver) were collected from a cohort of responders (N = 8) and non-responders (N = 8) based on RECIST v1.1 and examined by multiplex IHC and mRNA sequencing. RESULTS Treatment altered the transcriptional profile of metastatic lesions with a decrease in tumor cell proliferation independent of treatment response. The antiproliferative response was seen in both basal and classical PDA subtypes. PDA subtype was not associated with survival outcomes; instead, genes involved in immune activation distinguished responders from non-responders. Tumor response was associated with an increase in CD3+ and CD8+ T-cell infiltrates into metastatic lesions. A composite of decreased tumor proliferation in response to treatment and increased CD8 T-cell infiltration in metastatic lesions identified responders and associated with a favorable survival outcome. CONCLUSIONS Our findings suggest that inhibiting cancer cell proliferation alone in PDA is insufficient to produce tumor responses and support a role for tumor-extrinsic mechanisms, such as CD8+ T cells, which combine with the cancer cell proliferation index to define treatment outcomes.
Collapse
Affiliation(s)
- Gregory L. Beatty
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Devora Delman
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Authors contributed equally
| | - Jiayi Yu
- Newlink Genetics (Now LUMOS Pharmaceuticals), 2503 S Loop Dr. #5100, Ames, IA 50010
- Authors contributed equally
| | - Mingen Liu
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joey H. Li
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Liti Zhang
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jae W. Lee
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Renee B. Chang
- Abramson Cancer Center; University of Pennsylvania, Philadelphia, PA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nathan Bahary
- Department of Hematology-Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA
| | - Eugene P. Kennedy
- Newlink Genetics (Now LUMOS Pharmaceuticals), 2503 S Loop Dr. #5100, Ames, IA 50010
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO
| | - Gabriela R. Rossi
- Newlink Genetics (Now LUMOS Pharmaceuticals), 2503 S Loop Dr. #5100, Ames, IA 50010
| | | |
Collapse
|
94
|
Engler M, Albers D, Von Maltitz P, Groß R, Münch J, Cirstea IC. ACE2-EGFR-MAPK signaling contributes to SARS-CoV-2 infection. Life Sci Alliance 2023; 6:e202201880. [PMID: 37402592 PMCID: PMC10320016 DOI: 10.26508/lsa.202201880] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
SARS-CoV-2 triggered the most severe pandemic of recent times. To enter into a host cell, SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2). However, subsequent studies indicated that other cell membrane receptors may act as virus-binding partners. Among these receptors, the epidermal growth factor receptor (EGFR) was hypothesized not only as a spike protein binder, but also to be activated in response to SARS-CoV-2. In our study, we aim at dissecting EGFR activation and its major downstream signaling pathway, the mitogen-activated signaling pathway (MAPK), in SARS-CoV-2 infection. Here, we demonstrate the activation of EGFR-MAPK signaling axis by the SARS-CoV-2 spike protein and we identify a yet unknown cross talk between ACE2 and EGFR that regulated ACE2 abundance and EGFR activation and subcellular localization, respectively. By inhibiting the EGFR-MAPK activation, we observe a reduced infection with either spike-pseudotyped particles or authentic SARS-CoV-2, thus indicating that EGFR serves as a cofactor and the activation of EGFR-MAPK contributes to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Melanie Engler
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Pascal Von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | |
Collapse
|
95
|
Xing Y, Xue W, Teng Y, Jin Z, Tang X, Li Z, Hu Y, Wang R, Qian J. Raddeanin A promotes autophagy-induced apoptosis by inactivating PI3K/AKT/mTOR pathway in lung adenocarcinoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1987-1997. [PMID: 36882566 DOI: 10.1007/s00210-023-02447-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is the most common cancer in the world. Previous studies have shown that Raddeanin A (RA) exhibited distinct antitumor properties in gastric and colon cancer. This study aimed to investigate the pharmacological actions and intrinsic mechanisms of RA in NSCLC. Through the application of network pharmacology, the potential targets of RA for NSCLC therapy such as SRC, MAPK1, and STAT3 were excavated. Enrichment analyses showed that these targets were concerned with the regulation of cell death, regulation of MAPK cascade, Ras signaling pathway, and PI3K/AKT signaling pathway. Meanwhile, 13 targets of RA were identified as autophagy-related genes. Our experiment data showed that RA effectively inhibited proliferation and induced apoptosis in lung cancer cells A549. We also found that RA could induce autophagy simultaneously. Furthermore, the autophagy induced by RA had a synergistic effect with apoptosis and contributed to cell death. Additionally, RA could downregulate the activity of the PI3K/AKT/mTOR pathway. Generally, our results indicated the antitumor effect and underlying mechanisms of RA on apoptosis and autophagy in A549 cells, suggesting that RA could be used as an effective antineoplastic agent.
Collapse
Affiliation(s)
- Ying Xing
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Weiwei Xue
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yuhao Teng
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Zhichao Jin
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xiaolong Tang
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Suzhou Integrated Traditional Chinese and Western Medicine Hospital, Suzhou, China
| | - Zirui Li
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yue Hu
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ruiping Wang
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China.
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Jun Qian
- Department of Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, No.155, Hanzhong Road, Nanjing, 210029, China.
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
96
|
Elhariri A, Alhaj A, Ahn D, Sonbol MB, Bekaii-Saab T, Wu C, Rutenberg MS, Stauffer J, Starr J, Majeed U, Jones J, Borad M, Babiker H. Targeting KRAS in pancreatic adenocarcinoma: Progress in demystifying the holy grail. World J Clin Oncol 2023; 14:285-296. [PMID: 37700806 PMCID: PMC10494558 DOI: 10.5306/wjco.v14.i8.285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most challenging diseases, with a very poor 5-year overall survival of around 11.5%. Kirsten rat sarcoma virus (KRAS) mutation is seen in 90%-95% of PC patients and plays an important role in cancer cell proliferation, differentiation, metabolism, and survival, making it an essential mutation for targeted therapy. Despite extensive efforts in studying this oncogene, there has been little success in finding a drug to target this pathway, labelling it for decades as "undruggable". In this article we summarize some of the efforts made to target the KRAS pathway in PC, discuss the challenges, and shed light on promising clinical trials.
Collapse
Affiliation(s)
- Ahmed Elhariri
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Ahmed Alhaj
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Daniel Ahn
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Mohamad Bassam Sonbol
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Tanios Bekaii-Saab
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Christina Wu
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Michael Scott Rutenberg
- Department of Radiation-Oncology, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - John Stauffer
- Department of Surgical Oncology, Hepatopancreatobiliary Surgery, Mayo Clinic Florida, Jacksonville, FL 32224, United States
| | - Jason Starr
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Umair Majeed
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Jeremy Jones
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| | - Mitesh Borad
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Arizona, Mayo Clinic Cancer Center, Phoenix, AZ 85054, United States
| | - Hani Babiker
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Florida, Mayo Clinic Cancer Center, Jacksonville, FL 32224, United States
| |
Collapse
|
97
|
Sekiya S, Fukuda J, Yamamura R, Ooshio T, Satoh Y, Kosuge S, Sato R, Hatanaka KC, Hatanaka Y, Mitsuhashi T, Nakamura T, Matsuno Y, Hirano S, Sonoshita M. Drosophila Screening Identifies Dual Inhibition of MEK and AURKB as an Effective Therapy for Pancreatic Ductal Adenocarcinoma. Cancer Res 2023; 83:2704-2715. [PMID: 37378549 DOI: 10.1158/0008-5472.can-22-3762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
Significant progress has been made in understanding the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) by generating and using murine models. To accelerate drug discovery by identifying novel therapeutic targets on a systemic level, here we generated a Drosophila model mimicking the genetic signature in PDAC (KRAS, TP53, CDKN2A, and SMAD4 alterations), which is associated with the worst prognosis in patients. The '4-hit' flies displayed epithelial transformation and decreased survival. Comprehensive genetic screening of their entire kinome revealed kinases including MEK and AURKB as therapeutic targets. Consistently, a combination of the MEK inhibitor trametinib and the AURKB inhibitor BI-831266 suppressed the growth of human PDAC xenografts in mice. In patients with PDAC, the activity of AURKB was associated with poor prognosis. This fly-based platform provides an efficient whole-body approach that complements current methods for identifying therapeutic targets in PDAC. SIGNIFICANCE Development of a Drosophila model mimicking genetic alterations in human pancreatic ductal adenocarcinoma provides a tool for genetic screening that identifies MEK and AURKB inhibition as a potential treatment strategy.
Collapse
Affiliation(s)
- Sho Sekiya
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Junki Fukuda
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Takako Ooshio
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yusuke Satoh
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Kosuge
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Reo Sato
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Masahiro Sonoshita
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| |
Collapse
|
98
|
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, Lintas C, Tabolacci C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2023; 15:4026. [PMID: 37627054 PMCID: PMC10452867 DOI: 10.3390/cancers15164026] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer resulting from the malignant transformation of melanocytes. Recent therapeutic approaches, including targeted therapy and immunotherapy, have improved the prognosis and outcome of melanoma patients. BRAF is one of the most frequently mutated oncogenes recognised in melanoma. The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ERK (MAPK) signalling pathway. Therefore, mutated BRAF has become a useful target for molecular therapy and the use of BRAF kinase inhibitors has shown promising results. However, several resistance mechanisms invariably develop leading to therapeutic failure. The aim of this manuscript is to review the role of BRAF mutational status in the pathogenesis of melanoma and its impact on differentiation and inflammation. Moreover, this review focuses on the mechanisms responsible for resistance to targeted therapies in BRAF-mutated melanoma and provides an overview of circulating biomarkers including circulating tumour cells, circulating tumour DNA, and non-coding RNAs.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Bellenghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| |
Collapse
|
99
|
Yang J, Liu Y, Liu S. The role of epithelial-mesenchymal transition and autophagy in pancreatic ductal adenocarcinoma invasion. Cell Death Dis 2023; 14:506. [PMID: 37550301 PMCID: PMC10406904 DOI: 10.1038/s41419-023-06032-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/20/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Of all pancreatic cancer (PC) cases, approximately 90% are pancreatic ductal adenocarcinoma (PDAC), which progress rapidly due to its high degree of invasiveness and high metastatic potential. Epithelial-mesenchymal transition (EMT) is a prerequisite for cancer cell invasion and spread, and it is mediated by the specific cellular behaviors and the tumor microenvironment. Autophagy has long been a target of cancer therapy, and it has been considered to play a dual and contradictory role, particularly regarding EMT-mediated PDAC invasion. This review discusses the characteristics and the biological role of EMT and autophagy from a cellular perspective, explaining invasion as a survival behavior of PDAC, with the aim of providing novel insights into targeting EMT and autophagy to overcome PDAC invasion.
Collapse
Affiliation(s)
- Jian Yang
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang Province, P.R. China
| | - Ying Liu
- Department of Medical Oncology, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang Province, P.R. China
| | - Shi Liu
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang Province, P.R. China.
| |
Collapse
|
100
|
Huang Z, Kaller M, Hermeking H. CRISPR/Cas9-mediated inactivation of miR-34a and miR-34b/c in HCT116 colorectal cancer cells: comprehensive characterization after exposure to 5-FU reveals EMT and autophagy as key processes regulated by miR-34. Cell Death Differ 2023; 30:2017-2034. [PMID: 37488217 PMCID: PMC10406948 DOI: 10.1038/s41418-023-01193-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
The miR-34a and miR-34b/c encoding genes represent direct targets of the p53 transcription factor, and presumably mediate part of the tumor suppressive effects of p53. Here, we sought to determine their functional relevance by inactivating miR-34a and/or miR-34b/c using a CRISPR/Cas9 approach in the colorectal cancer (CRC) cell line HCT116. Concomitant deletion of miR-34a and miR-34b/c resulted in significantly reduced suppression of proliferation after p53 activation, enhanced migration, invasion and EMT, as well as reduced sensitivity to chemotherapeutics, increased stress-induced autophagic flux, decreased apoptosis and upregulation of autophagy-related genes after 5-FU treatment. However, inactivation of singular miR-34a or miR-34b/c had little effects on the aforementioned processes. RNA-Seq analysis revealed that concomitant deletion of miR-34a/b/c caused EMT signature enrichment, impaired gene repression by the p53-DREAM pathway and elevated autophagy after 5-FU treatment. A gene signature comprised of mRNAs significantly upregulated after combined inactivation of miR-34a and miR-34b/c showed a significant association with the invasive colon cancer subtype CMS4 and poor overall survival in two CRC patient cohorts, and with 5-FU resistance in CRC cell lines. In miR-34a/b/c-deficient cells the upregulated miR-34 target FOXM1 directly induced p62 and ATG9A, which increased autophagy and consequently attenuated apoptosis and rendered the miR-34a/b/c-KO cells more resistant to 5-FU. Inhibition of autophagy by depletion of ATG9A or chloroquine re-sensitized miR-34a/b/c-deficient HCT116 cells to 5-FU. In summary, our findings show a complementary role of miR-34a and miR-34b/c in the regulation of EMT and autophagy which may be relevant for CRC therapy in the future.
Collapse
Affiliation(s)
- Zekai Huang
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Markus Kaller
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, D-80336, Munich, Germany.
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany.
| |
Collapse
|