1
|
Boka HJ, Engel RM, Georges C, McMurrick PJ, Abud HE. Does side matter? Deciphering mechanisms that underpin side-dependent pathogenesis and therapy response in colorectal cancer. Mol Cancer 2025; 24:130. [PMID: 40312719 PMCID: PMC12046799 DOI: 10.1186/s12943-025-02327-5] [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: 02/20/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Colorectal cancer (CRC) is stratified by heterogeneity between disease sites, with proximal right-sided CRC (RCRC) multifactorial in its distinction from distal left-sided CRC (LCRC). Notably, right-sided tumors are associated with aggressive disease characteristics which culminate in poor clinical outcomes for these patients. While factors such as mutational profile and patterns of metastasis have been suggested to contribute to differences in therapy response, the exact mechanisms through which RCRC resists effective treatment have yet to be elucidated. In response, recent analyzes, including those utilizing whole genome sequencing, transcriptional profiling, and single-cell analyses, have demonstrated that key molecular differences exist between disease sites, with differentially expressed genes spanning a diverse range of cellular functions. Here, we review and contextualize the most recent data on molecular biomarkers found to exhibit discordance between RCRC and LCRC, and highlight candidates for further investigation, including those which present promise for future clinical application. Given the present disparity in survival outcomes for RCRC patients, we expect the prognostic biomarkers presented in our review to be useful in establishing future directions for the side-specific treatment of CRC.
Collapse
Affiliation(s)
- Harrison J Boka
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Rebekah M Engel
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Christine Georges
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Paul J McMurrick
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Department of Surgery, Cabrini Monash University, Cabrini Hospital, Malvern, VIC, 3144, Australia.
| |
Collapse
|
2
|
Marx C, Qing X, Gong Y, Kirkpatrick J, Siniuk K, Beznoussenko GV, Kidiyoor G, Kirtay M, Buder K, Koch P, Westermann M, Bruhn C, Brown E, Xu X, Foiani M, Wang ZQ. DNA damage response regulator ATR licenses PINK1-mediated mitophagy. Nucleic Acids Res 2025; 53:gkaf178. [PMID: 40105243 PMCID: PMC11920799 DOI: 10.1093/nar/gkaf178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Defective DNA damage response (DDR) and mitochondrial dysfunction are a major etiology of tissue impairment and aging. Mitochondrial autophagy (mitophagy) is a mitochondrial quality control (MQC) mechanism to selectively eliminate dysfunctional mitochondria. ATR (ataxia-telangiectasia and Rad3-related) is a key DDR regulator playing a pivotal role in DNA replication stress response and genomic stability. Paradoxically, the human Seckel syndrome caused by ATR mutations exhibits premature aging and neuropathies, suggesting a role of ATR in nonreplicating tissues. Here, we report a previously unknown yet direct role of ATR at mitochondria. We find that ATR and PINK1 (PTEN-induced kinase 1) dock at the mitochondrial translocase TOM/TIM complex, where ATR interacts directly with and thereby stabilizes PINK1. ATR deletion silences mitophagy initiation thereby altering oxidative phosphorylation functionality resulting in reactive oxygen species overproduction that attack cytosolic macromolecules, in both cells and brain tissues, prior to nuclear DNA. This study discloses ATR as an integrated component of the PINK1-mediated MQC program to ensure mitochondrial fitness. Together with its DDR function, ATR safeguards mitochondrial and genomic integrity under physiological and genotoxic conditions.
Collapse
Affiliation(s)
- Christian Marx
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
- Center for Pandemic Vaccines and Therapeutics (ZEPAI), Paul Ehrlich Institute (PEI), Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Xiaobing Qing
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Yamin Gong
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
- Faculty of Basic Medicine, Shenzhen University Medical School, 518055 Shenzhen, China
| | - Joanna Kirkpatrick
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Kanstantsin Siniuk
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | | | | | - Murat Kirtay
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Katrin Buder
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Philipp Koch
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Christopher Bruhn
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Eric J Brown
- Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, United States
| | - Xingzhi Xu
- Faculty of Basic Medicine, Shenzhen University Medical School, 518055 Shenzhen, China
| | - Marco Foiani
- IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller-University of Jena, Bachstraße 18k, 07743 Jena, Germany
- State Key Laboratory of Microbial Technology, Shandong University, 266237 Qingdao, China
| |
Collapse
|
3
|
Tavakoli N, Fong EJ, Coleman A, Huang YK, Bigger M, Doche ME, Kim S, Lenz HJ, Graham NA, Macklin P, Finley SD, Mumenthaler SM. Merging metabolic modeling and imaging for screening therapeutic targets in colorectal cancer. NPJ Syst Biol Appl 2025; 11:12. [PMID: 39875420 PMCID: PMC11775273 DOI: 10.1038/s41540-025-00494-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in metabolic reprogramming and are well-established contributors to drug resistance in colorectal cancer (CRC). To exploit this metabolic crosstalk, we integrated a systems biology approach that identified key metabolic targets in a data-driven method and validated them experimentally. This process involved a novel machine learning-based method to computationally screen, in a high-throughput manner, the effects of enzyme perturbations predicted by a computational model of CRC metabolism. This approach reveals the network-wide effects of metabolic perturbations. Our results highlighted hexokinase (HK) as a crucial target, which subsequently became our focus for experimental validation using patient-derived tumor organoids (PDTOs). Through metabolic imaging and viability assays, we found that PDTOs cultured in CAF-conditioned media exhibited increased sensitivity to HK inhibition, confirming the model predictions. Our approach emphasizes the critical role of integrating computational and experimental techniques in exploring and exploiting CRC-CAF crosstalk.
Collapse
Affiliation(s)
- Niki Tavakoli
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Emma J Fong
- Ellison Medical Institute, Los Angeles, CA, 90064, USA
| | | | - Yu-Kai Huang
- Ellison Medical Institute, Los Angeles, CA, 90064, USA
| | - Mathias Bigger
- Ellison Medical Institute, Los Angeles, CA, 90064, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | | | - Seungil Kim
- Ellison Medical Institute, Los Angeles, CA, 90064, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 46202, USA
| | - Stacey D Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Shannon M Mumenthaler
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
- Ellison Medical Institute, Los Angeles, CA, 90064, USA.
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
4
|
Tavakoli N, Fong EJ, Coleman A, Huang YK, Bigger M, Doche ME, Kim S, Lenz HJ, Graham NA, Macklin P, Finley SD, Mumenthaler SM. Merging Metabolic Modeling and Imaging for Screening Therapeutic Targets in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595756. [PMID: 38826317 PMCID: PMC11142224 DOI: 10.1101/2024.05.24.595756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in metabolic reprogramming and are well-established contributors to drug resistance in colorectal cancer (CRC). To exploit this metabolic crosstalk, we integrated a systems biology approach that identified key metabolic targets in a data-driven method and validated them experimentally. This process involved a novel machine learning-based method to computationally screen, in a high-throughput manner, the effects of enzyme perturbations predicted by a computational model of CRC metabolism. This approach reveals the network-wide effects of metabolic perturbations. Our results highlighted hexokinase (HK) as a crucial target, which subsequently became our focus for experimental validation using patient-derived tumor organoids (PDTOs). Through metabolic imaging and viability assays, we found that PDTOs cultured in CAF-conditioned media exhibited increased sensitivity to HK inhibition, confirming the model predictions. Our approach emphasizes the critical role of integrating computational and experimental techniques in exploring and exploiting CRC-CAF crosstalk.
Collapse
|
5
|
Fan K, Wang Y, Bian J, Sun Y, Dou J, Pan J, Yu Y. Study on the effects of rapamycin and the mTORC1/2 dual inhibitor OSI-027 on the metabolism of colon cancer cells based on UPLC-MS/MS metabolomics. Invest New Drugs 2024; 42:418-427. [PMID: 38916794 DOI: 10.1007/s10637-024-01438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/16/2024] [Indexed: 06/26/2024]
Abstract
mTORC1/2 dual inhibitors may be more effective than mTORC1 inhibitor rapamycin. However, their metabolic impacts on colon cancer cells remain unexplored. We conducted a comparative analysis of the anti-proliferative effects of rapamycin and the novel OSI-027 in colon cancer cells HCT-116, evaluating their metabolic influences through ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Our results demonstrate that OSI-027 more effectively inhibits colon cancer cell proliferation than rapamycin. Additionally, we identified nearly 600 metabolites from the spectra, revealing significant differences in metabolic patterns between cells treated with OSI-027 and rapamycin. Through VIP value screening, we pinpointed crucial metabolites contributing to these distinctions. For inhibiting glycolysis and reducing glucose consumption, OSI-027 was likely to be more potent than rapamycin. For amino acids metabolism, although OSI-027 has a broad effect as rapamycin, their effects in degrees were not exactly the same. These findings address the knowledge gap regarding mTORC1/2 dual inhibitors and lay a foundation for their further development and research.
Collapse
Affiliation(s)
- Kai Fan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Yueyuan Wang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Jiangyujing Bian
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
- College of Pharmacy, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yewen Sun
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
- College of Pharmacy, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Jiaqi Dou
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China.
| | - Yunli Yu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
Zhang Y, Yan H, Wei Y, Wei X. Decoding mitochondria's role in immunity and cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189107. [PMID: 38734035 DOI: 10.1016/j.bbcan.2024.189107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The functions of mitochondria, including energy production and biomolecule synthesis, have been known for a long time. Given the rising incidence of cancer, the role of mitochondria in cancer has become increasingly popular. Activated by components released by mitochondria, various pathways interact with each other to induce immune responses to protect organisms from attack. However, mitochondria play dual roles in the progression of cancer. Abnormalities in proteins, which are the elementary structures of mitochondria, are closely linked with oncogenesis. Both the aberrant accumulation of intermediates and mutations in enzymes result in the generation and progression of cancer. Therefore, targeting mitochondria to treat cancer may be a new strategy. Several drugs aimed at inhibiting mutated enzymes and accumulated intermediates have been tested clinically. Here, we discuss the current understanding of mitochondria in cancer and the interactions between mitochondrial functions, immune responses, and oncogenesis. Furthermore, we discuss mitochondria as hopeful targets for cancer therapy, providing insights into the progression of future therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Hong Yan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| |
Collapse
|
7
|
Zhang Y, Hao M, Yang X, Zhang S, Han J, Wang Z, Chen HN. Reactive oxygen species in colorectal cancer adjuvant therapies. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166922. [PMID: 37898425 DOI: 10.1016/j.bbadis.2023.166922] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Colorectal cancer (CRC), a prevalent global malignancy, often necessitates adjuvant therapies such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy to mitigate tumor burden in advanced stages. The efficacy of these therapies is significantly influenced by reactive oxygen species (ROS). Previous research underscores the pivotal role of ROS in gut pathology, targeted therapy, and drug resistance. ROS-mediated CRC adjuvant therapies encompass a myriad of mechanisms, including cell death and proliferation, survival and cell cycle, DNA damage, metabolic reprogramming, and angiogenesis. Preliminary clinical trials have begun to unveil the potential of ROS-manipulating therapy in enhancing CRC adjuvant therapies. This review aims to provide a comprehensive synthesis of studies exploring the role of ROS in CRC adjuvant therapies.
Collapse
Affiliation(s)
- Yang Zhang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengqiu Hao
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Su Zhang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziqiang Wang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hai-Ning Chen
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Pendleton KE, Wang K, Echeverria GV. Rewiring of mitochondrial metabolism in therapy-resistant cancers: permanent and plastic adaptations. Front Cell Dev Biol 2023; 11:1254313. [PMID: 37779896 PMCID: PMC10534013 DOI: 10.3389/fcell.2023.1254313] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Deregulation of tumor cell metabolism is widely recognized as a "hallmark of cancer." Many of the selective pressures encountered by tumor cells, such as exposure to anticancer therapies, navigation of the metastatic cascade, and communication with the tumor microenvironment, can elicit further rewiring of tumor cell metabolism. Furthermore, phenotypic plasticity has been recently appreciated as an emerging "hallmark of cancer." Mitochondria are dynamic organelles and central hubs of metabolism whose roles in cancers have been a major focus of numerous studies. Importantly, therapeutic approaches targeting mitochondria are being developed. Interestingly, both plastic (i.e., reversible) and permanent (i.e., stable) metabolic adaptations have been observed following exposure to anticancer therapeutics. Understanding the plastic or permanent nature of these mechanisms is of crucial importance for devising the initiation, duration, and sequential nature of metabolism-targeting therapies. In this review, we compare permanent and plastic mitochondrial mechanisms driving therapy resistance. We also discuss experimental models of therapy-induced metabolic adaptation, therapeutic implications for targeting permanent and plastic metabolic states, and clinical implications of metabolic adaptations. While the plasticity of metabolic adaptations can make effective therapeutic treatment challenging, understanding the mechanisms behind these plastic phenotypes may lead to promising clinical interventions that will ultimately lead to better overall care for cancer patients.
Collapse
Affiliation(s)
- Katherine E. Pendleton
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Karen Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Gloria V. Echeverria
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
9
|
Moretton A, Kourtis S, Gañez Zapater A, Calabrò C, Espinar Calvo ML, Fontaine F, Darai E, Abad Cortel E, Block S, Pascual‐Reguant L, Pardo‐Lorente N, Ghose R, Vander Heiden MG, Janic A, Müller AC, Loizou JI, Sdelci S. A metabolic map of the DNA damage response identifies PRDX1 in the control of nuclear ROS scavenging and aspartate availability. Mol Syst Biol 2023; 19:e11267. [PMID: 37259925 PMCID: PMC10333845 DOI: 10.15252/msb.202211267] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
While cellular metabolism impacts the DNA damage response, a systematic understanding of the metabolic requirements that are crucial for DNA damage repair has yet to be achieved. Here, we investigate the metabolic enzymes and processes that are essential for the resolution of DNA damage. By integrating functional genomics with chromatin proteomics and metabolomics, we provide a detailed description of the interplay between cellular metabolism and the DNA damage response. Further analysis identified that Peroxiredoxin 1, PRDX1, contributes to the DNA damage repair. During the DNA damage response, PRDX1 translocates to the nucleus where it reduces DNA damage-induced nuclear reactive oxygen species. Moreover, PRDX1 loss lowers aspartate availability, which is required for the DNA damage-induced upregulation of de novo nucleotide synthesis. In the absence of PRDX1, cells accumulate replication stress and DNA damage, leading to proliferation defects that are exacerbated in the presence of etoposide, thus revealing a role for PRDX1 as a DNA damage surveillance factor.
Collapse
Affiliation(s)
- Amandine Moretton
- Center for Cancer Research, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Savvas Kourtis
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Antoni Gañez Zapater
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Chiara Calabrò
- Center for Cancer Research, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | | | - Frédéric Fontaine
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Evangelia Darai
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Etna Abad Cortel
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Samuel Block
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Laura Pascual‐Reguant
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Natalia Pardo‐Lorente
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Ritobrata Ghose
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- Dana‐Farber Cancer InstituteBostonMAUSA
| | - Ana Janic
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - André C Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Joanna I Loizou
- Center for Cancer Research, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Sara Sdelci
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| |
Collapse
|
10
|
Chiu PF, Chang CK, Huang PS, Lin YY, Lin CS, Yang HY, Hsu LC, Yu LCH, Liang PH. Design, Synthesis, and Evaluation of Glucose Transporter Inhibitor-SN38 Conjugates for Targeting Colorectal Cancer. J Med Chem 2023. [PMID: 37413981 DOI: 10.1021/acs.jmedchem.3c00476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Irinotecan (1), a prodrug of SN38 (2) approved by the US Food and Drug Administration for treating colorectal cancer, lacks specificity and causes many side effects. To increase the selectivity and therapeutic efficacy of this drug, we designed and synthesized conjugates of SN38 and glucose transporter inhibitors (phlorizin (5) or phloretin (6)), which could be hydrolyzed by glutathione or cathepsin to release SN38 in the tumor microenvironment, as a proof of concept. These conjugates (8, 9, and 10) displayed better antitumor efficacy with lower systemic exposure to SN38 in an orthotopic colorectal cancer mouse model compared with irinotecan at the same dosage. Further, no major adverse effects of the conjugates were observed during treatment. Biodistribution studies showed that conjugate 10 could induce higher concentrations of free SN38 in tumor tissues than irinotecan at the same dosage. Thus, the developed conjugates exhibit potential for treating colorectal cancer.
Collapse
Affiliation(s)
- Pei-Fang Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chun-Kai Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Pin-Shuo Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - You-Yu Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chung-Shun Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hui-Yi Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- The Genomics Research Center, Academia Sinica, Taipei 128, Taiwan
| |
Collapse
|
11
|
Napolitano S, Woods M, Lee HM, De Falco V, Martini G, Della Corte CM, Martinelli E, Famiglietti V, Ciardiello D, Anderson A, Fowlkes NW, Villareal OE, Sorokin A, Kanikarla P, Coker O, Morris V, Altucci L, Tabernero J, Troiani T, Ciardiello F, Kopetz S. Antitumor Efficacy of Dual Blockade with Encorafenib + Cetuximab in Combination with Chemotherapy in Human BRAFV600E-Mutant Colorectal Cancer. Clin Cancer Res 2023; 29:2299-2309. [PMID: 37040395 PMCID: PMC10261917 DOI: 10.1158/1078-0432.ccr-22-3894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/14/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
PURPOSE Encorafenib + cetuximab (E+C) is an effective therapeutic option in chemorefractory BRAFV600E metastatic colorectal cancer (mCRC). However, there is a need to improve the efficacy of this molecular-targeted therapy and evaluate regimens suitable for untreated BRAFV600E in patients with mCRC. EXPERIMENTAL DESIGN We performed a series of in vivo studies using BRAFV600E mCRC tumor xenografts. Mice were randomized to receive 5-fluoruracil (5-FU), irinotecan, or oxaliplatin regimens (FOLFIRI or FOLFOX), (E+C) or the combination. Patients received long-term treatment until disease progression, with deescalation strategies used to mimic maintenance therapy. Transcriptomic changes after progression on cytotoxic chemotherapy or targeted therapy were assessed. RESULTS Antitumor activity of either FOLFIRI or E+C was better as first-line treatment as compared with second-line, with partial cross-resistance seen between a cytotoxic regimen and targeted therapy with an average 62% loss of efficacy for FOLFIRI after E+C and a 45% loss of efficacy of E+C after FOLFIRI (P < 0.001 for both). FOLFIRI-treated models had upregulation of epithelial-mesenchymal transition (EMT) and MAPK pathway activation, where E+C treated models had suppressed MAPK signaling. In contrast, with chemotherapy with E+C, EMT and MAPK signaling remained suppressed. FOLFOX or FOLFIRI, each in combination with E+C, were the most active first-line treatments as compared with E+C or to chemotherapy alone. Furthermore, FOLFOX in combination with E+C as first-line induction therapy, followed by E+C ± 5-FU as maintenance therapy, was the most effective strategy for long-term disease control. CONCLUSIONS These results support the combination of cytotoxic chemotherapy and molecular-targeted therapy as a promising therapeutic approach in the first-line treatment of BRAFV600E mCRC.
Collapse
Affiliation(s)
- Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Melanie Woods
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hey Min Lee
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vincenzo De Falco
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Giulia Martini
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Carminia Maria Della Corte
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Vincenzo Famiglietti
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Amanda Anderson
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Natalie Wall Fowlkes
- Veterinary Medicine & Surgery Department, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Oscar Eduardo Villareal
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexey Sorokin
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Preeti Kanikarla
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Olu Coker
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Van Morris
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lucia Altucci
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Josep Tabernero
- Medical Oncology Department, Vall d' Hebron Hospital Campus, Barcelona, Spain
- Institute of Oncology, University of Vic/Central University of Catalonia, Barcelona, Spain
- Oncology Institute of Barcelona-Quironsalud, Biomedical Research Center in Cancer, Barcelona, Spain
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Scott Kopetz
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|