1
|
Hu Y, He X, Li S, Zhang T, Liao J, Xu N, Yuan Y, Wang Q, Chen Z, Huang J, Ma L. Hit to lead optimization of isopentenyl chalcones as novel MTHFD2 inhibitors for cancer treatment: design, synthesis, in-vitro, in-vivo and in-silico studies. Eur J Med Chem 2025; 292:117703. [PMID: 40334504 DOI: 10.1016/j.ejmech.2025.117703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/20/2025] [Accepted: 04/27/2025] [Indexed: 05/09/2025]
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) plays a key role in one-carbon metabolism, as it is highly upregulated in cancer cells while exhibiting minimal expression in healthy adult tissues. Consequently, MTHFD2 is regarded as a promising target for cancer therapies. In this study, a series of isopentenyl chalcones, based on hit compound sophoradin, were designed and synthesized by computer-aided drug design. Preliminary structure-activities relationship revealed the great significance of chalcone scaffold and isopentenyl groups. The optimized compound 41, with an isopentenyl group and three hydroxyl groups, demonstrated remarkable activity and high selectivity in enzymatic assays (MTHFD1 IC50 = 19.05 ± 7.10 μM, MTHFD2 IC50 = 1.46 ± 0.28 μM, SI = 13). The cellular thermal shift assay implied that 41 could directly bind to MTHFD2. In vitro, compound 41 dramatically promoted intracellular ROS accumulation, and exhibited potent antiproliferative activity against lung cancer cells H1299 with low toxicity to BEAS-2B cells. Furthermore, 41 also demonstrated considerable anti-lung cancer efficacy in a mouse xenograft model and favorable pharmacokinetic properties without significant abnormalities in major organs. This work enriches the structure-activity relationship of MTHFD2 inhibitors and provides a potential candidate for cancer treatment.
Collapse
Affiliation(s)
- Yingjie Hu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Xiangli He
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Shuhui Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Tingting Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Jingjing Liao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Ning Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | - Qi Wang
- Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| | - Zhuo Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| |
Collapse
|
2
|
Liu S, Wang X, Zhao L, Zhang L, Song Y. MTHFD2: A significant mitochondrial metabolic enzyme and a novel target for anticancer therapy. Drug Discov Today 2025; 30:104372. [PMID: 40348077 DOI: 10.1016/j.drudis.2025.104372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a crucial mitochondrial enzyme that operates within the folate one-carbon metabolic pathway. In recent years, it has been discovered that its expression is upregulated in numerous tumors and is correlated with the onset and progression of tumors, as well as poor prognosis. In contrast to its isoenzymes, it is overexpressed in tumors and is either expressed at low levels or not expressed at all in normal tissues. Consequently, it has received extensive attention and has been proposed as a novel anticancer target. In this paper, we review the functions of MTHFD2 in tumors, its regulatory mechanisms, and research progress on MTHFD2 inhibitors. Additionally, we provide insights into future research directions and the design and development of inhibitors for MTHFD2.
Collapse
Affiliation(s)
- Shuyu Liu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Xiaoke Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei 071002, China
| | - Liye Zhao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100091, China
| | - Yali Song
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, Hebei 071002, China.
| |
Collapse
|
3
|
Chang HH, Lee LC, Hsu T, Peng YH, Huang CH, Yeh TK, Lu CT, Huang ZT, Hsueh CC, Kung FC, Lin LM, Huang YC, Wang YH, Li LH, Tang YC, Chang L, Hsieh CC, Jiaang WT, Kuo CC, Wu SY. Development of Potent and Selective Inhibitors of Methylenetetrahydrofolate Dehydrogenase 2 for Targeting Acute Myeloid Leukemia: SAR, Structural Insights, and Biological Characterization. J Med Chem 2024; 67:21106-21125. [PMID: 39591507 DOI: 10.1021/acs.jmedchem.4c01775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Methylenetetrahydrofolate dehydrogenase/cyclohydrolase 2 (MTHFD2), a pivotal mitochondrial enzyme in one-carbon metabolism, is significantly upregulated in various cancers but minimally expressed in normal proliferating cells. In contrast, MTHFD1, which performs similar functions, is predominantly expressed in normal cells. Therefore, targeting MTHFD2 with selective inhibitors holds promise for a broader therapeutic window with reduced toxicity and fewer side effects. This study identified selective 2,4-diamino-6-oxo-1,6-dihydropyrimidin-5-yl ureido-based derivatives through systematic chemical modifications and SAR studies. Structural biology investigations revealed substitutions in the phenyl ring and tail region modulate potency and selectivity toward MTHFD2. Additionally, a comprehensive cell screening platform revealed acute myeloid leukemia cells with FLT3 internal tandem duplication mutations are particularly sensitive to these inhibitors. Furthermore, synergistic effects were observed when combining potential compounds with Alimta. Compound 16e emerged as a leading candidate, demonstrating superior inhibition and selectivity for MTHFD2, favorable pharmacokinetics, and potent antitumor efficacy in MOLM-14 xenograft models.
Collapse
Affiliation(s)
- Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Lung-Chun Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Tsu Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Yi-Hui Peng
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Chih-Hsiang Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Cheng-Tai Lu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Zih-Ting Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Ching-Cheng Hsueh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Fang-Chun Kung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Li-Mei Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Yi-Hsin Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Li-Hsuan Li
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan, Republic of China
| | - Ya-Chu Tang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Ling Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Chih-Chien Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | - Su-Ying Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| |
Collapse
|
4
|
Hernández-Ayala LF, Guzmán-López EG, Pérez-González A, Reina M, Galano A. Molecular Insights on Coffee Components as Chemical Antioxidants. J MEX CHEM SOC 2024; 68:888-969. [DOI: 10.29356/jmcs.v68i4.2238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Coffee is not only a delicious beverage but also an important dietary source of natural antioxidants. We live in a world where it is impossible to avoid pollution, stress, food additives, radiation, and other sources of oxidants that eventually lead to severe health disorders. Fortunately, there are chemicals in our diet that counteract the hazards posed by the reactive species that trigger oxidative stress. They are usually referred to as antioxidants; some of them can be versatile compounds that exert such a role in many ways. This review summarizes, from a chemical point of view, the antioxidant effects of relevant molecules found in coffee. Their mechanisms of action, trends in activity, and the influence of media and pH in aqueous solutions, are analyzed. Structure-activity relationships are discussed, and the protective roles of these compounds are examined. A particular section is devoted to derivatives of some coffee components, and another one to their bioactivity. The data used in the analysis come from theoretical and computational protocols, which have been proven to be very useful in this context. Hopefully, the information provided here will pro-mote further investigations into the amazing chemistry contained in our morning coffee cup.
Resumen. El café no solo es una bebida deliciosa, sino también una importante fuente dietética de antioxidantes naturales. Vivimos en un mundo donde es imposible evitar la contaminación, el estrés, los aditivos alimentarios, la radiación y otras fuentes de oxidantes que eventualmente conducen a trastornos de salud graves. Afortunadamente, existen sustancias químicas en nuestra dieta que contrarrestan los peligros planteados por las especies reactivas que desencadenan el estrés oxidativo. Por lo general, se les denomina antioxidantes; algunos de ellos pueden ser compuestos versátiles que ejercen dicho papel de muchas maneras. Este artículo de revisión resume, desde un punto de vista químico, los efectos antioxidantes de moléculas relevantes encontradas en el café. Se analizan sus mecanismos de acción, tendencias en la actividad y la influencia del medio y el pH en soluciones acuosas. Se discuten las relaciones estructura-actividad, y se examinan los roles protectores de estos compuestos. Se dedica una sección particular a los derivados de algunos componentes del café, y otra a su bioactividad. Los datos utilizados en el análisis provienen de protocolos teóricos y computacionales, que han demostrado ser muy útiles en este contexto. Se espera que la información proporcionada aquí promueva investigaciones futuras sobre la química contenida en nuestra taza de café matutina.
Collapse
|
5
|
Jha V, Eriksson LA. Selectivity analysis of diaminopyrimidine-based inhibitors of MTHFD1, MTHFD2 and MTHFD2L. Sci Rep 2024; 14:21073. [PMID: 39256448 PMCID: PMC11387627 DOI: 10.1038/s41598-024-71879-1] [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: 05/07/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
The mitochondrial enzyme methylenetetrahydrofolate dehydrogenase (MTHFD2) is involved in purine and thymidine synthesis via 1C metabolism. MTHFD2 is exclusively overexpressed in cancer cells but absent in most healthy adult human tissues. However, the two close homologs of MTHFD2 known as MTHFD1 and MTHFD2L are expressed in healthy adult human tissues and share a great structural resemblance to MTHFD2 with 54% and 89% sequence similarity, respectively. It is therefore notably challenging to find selective inhibitors of MTHFD2 due to the structural similarity, in particular protein binding site similarity with MTHFD1 and MTHFD2L. Tricyclic coumarin-based compounds (substrate site binders) and xanthine derivatives (allosteric site binders) are the only selective inhibitors of MTHFD2 reported till date. Nanomolar potent diaminopyrimidine-based inhibitors of MTHFD2 have been reported recently, however, they also demonstrate significant inhibitory activities against MTHFD1 and MTHFD2L. In this study, we have employed extensive computational modeling involving molecular docking and molecular dynamics simulations in order to investigate the binding modes and key interactions of diaminopyrimidine-based inhibitors at the substrate binding sites of MTHFD1, MTHFD2 and MTHFD2L, and compare with the tricyclic coumarin-based selective MTHFD2 inhibitor. The outcomes of our study provide significant insights into desirable and undesirable structural elements for rational structure-based design of new and selective inhibitors of MTHFD2 against cancer.
Collapse
Affiliation(s)
- Vibhu Jha
- Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30, Göteborg, Sweden
- School of Pharmacy and Medical Sciences, Faculty of Life Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, BD71DP, UK
| | - Leif A Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30, Göteborg, Sweden.
| |
Collapse
|
6
|
Lee Y, Vousden KH, Hennequart M. Cycling back to folate metabolism in cancer. NATURE CANCER 2024; 5:701-715. [PMID: 38698089 PMCID: PMC7616045 DOI: 10.1038/s43018-024-00739-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/30/2024] [Indexed: 05/05/2024]
Abstract
Metabolic changes contribute to cancer initiation and progression through effects on cancer cells, the tumor microenvironment and whole-body metabolism. Alterations in serine metabolism and the control of one-carbon cycles have emerged as critical for the development of many tumor types. In this Review, we focus on the mitochondrial folate cycle. We discuss recent evidence that, in addition to supporting nucleotide synthesis, mitochondrial folate metabolism also contributes to metastasis through support of antioxidant defense, mitochondrial protein synthesis and the overflow of excess formate. These observations offer potential therapeutic opportunities, including the modulation of formate metabolism through dietary interventions and the use of circulating folate cycle metabolites as biomarkers for cancer detection.
Collapse
Affiliation(s)
| | | | - Marc Hennequart
- The Francis Crick Institute, London, UK
- Namur Research Institute for Life Sciences (NARILIS), Molecular Physiology Unit (URPHYM), University of Namur, Namur, Belgium
| |
Collapse
|
7
|
Rana N, Patel D, Parmar M, Mukherjee N, Jha PC, Manhas A. Targeting allosteric binding site in methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) to identify natural product inhibitors via structure-based computational approach. Sci Rep 2023; 13:18090. [PMID: 37872243 PMCID: PMC10593809 DOI: 10.1038/s41598-023-45175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
Cancer has been viewed as one of the deadliest diseases worldwide. Among various types of cancer, breast cancer is the most common type of cancer in women. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a promising druggable target and is overexpressed in cancerous cells, like, breast cancer. We conducted structure-based modeling on the allosteric site of the enzyme. Targeting the allosteric site avoids the problem of drug resistance. Pharmacophore modeling, molecular docking, HYDE assessment, drug-likeness, ADMET predictions, simulations, and free-energy calculations were performed. The RMSD, RMSF, RoG, SASA, and Hydrogen-bonding studies showed that seven candidates displayed stable behaviour. As per the literature, average superimposed simulated structures revealed a similar protein conformational change in the αE'-βf' loop, causing its displacement away from the allosteric site. The MM-PBSA showed tight binding of six compounds with the allosteric pocket. The effect of inhibitors interacting in the allosteric site causes a decrease in the binding energy of J49 (active-site inhibitor), suggesting the effect of allosteric binding. The PCA and FEL analysis revealed the significance of the docked compounds in the stable behaviour of the complexes. The outcome can contribute to the development of potential natural products with drug-like properties that can inhibit the MTHFD2 enzyme.
Collapse
Affiliation(s)
- Nisarg Rana
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, India
| | - Dhaval Patel
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gandhinagar, India
| | - Meet Parmar
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gandhinagar, India
| | - Nandini Mukherjee
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, India
| | - Prakash C Jha
- School of Applied Material Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Anu Manhas
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, India.
| |
Collapse
|
8
|
Jha V, Eriksson LA. Binding Modes of Xanthine-Derived Selective Allosteric Site Inhibitors of MTHFD2. ChemistryOpen 2023; 12:e202300052. [PMID: 37129313 PMCID: PMC10152887 DOI: 10.1002/open.202300052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/17/2023] [Indexed: 05/03/2023] Open
Abstract
Methylenetetrahydrofolate dehydrogenase (MTHFD2) is a mitochondrial enzyme involved in 1 C metabolism that is upregulated in various cancer cells, but absent in normal proliferating cells. Xanthine derivatives are the first selective inhibitors of MTHFD2 which bind to its allosteric site. Xanthine derivatives (including the co-crystallized inhibitors) were herein interrogated by molecular/induced-fit docking, MM-GBSA binding free energy calculations and molecular dynamics simulations in both MTHFD2 and MTHFD1 (a close homolog expressed in healthy cells). The gained insights from our in silico protocol allowed us to study binding mode, key protein-ligand interactions and dynamic movement of the allosteric inhibitors, correlating with their experimental binding affinities, biological activities and selectivity for MTHFD2. The reported conformational changes with MTHFD2 upon binding of xanthine derivatives were furthermore evaluated and confirmed by RMSF analyses of the MD simulation trajectories. The results reported herein are expected to benefit in the rational design of selective MTHFD2 allosteric inhibitors.
Collapse
Affiliation(s)
- Vibhu Jha
- Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9c, 405 30, Göteborg, Sweden
| | - Leif A Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9c, 405 30, Göteborg, Sweden
| |
Collapse
|
9
|
Jha V, Holmelin FL, Eriksson LA. Binding Analysis and Structure-Based Design of Tricyclic Coumarin-Derived MTHFD2 Inhibitors as Anticancer Agents: Insights from Computational Modeling. ACS OMEGA 2023; 8:14440-14458. [PMID: 37125100 PMCID: PMC10134251 DOI: 10.1021/acsomega.2c08025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Unfolded protein response (UPR)-dependent metabolic reprogramming diverts metabolites from glycolysis to mitochondrial 1C metabolism, highlighting pharmacological resistance to folate drugs and overexpression of certain enzymes. Methylenetetrahydrofolate dehydrogenase (MTHFD2) is a mitochondrial enzyme that plays a key role in 1C metabolism in purine and thymidine synthesis and is exclusively overexpressed in cancer cells but absent in most healthy adult human tissues. To the best of our knowledge, tricyclic coumarin-based compounds (substrate site binders) and xanthine derivatives (allosteric site binders) are the only selective inhibitors of MTHFD2 reported until the present date. The current study aims at the investigation of the available structural data of MTHFD2 in complex with potent and selective inhibitors that occupy the substrate binding site, further providing insights into binding mode, key protein-ligand interactions, and conformational dynamics, that correspond to the experimental binding affinities and biological activities. In addition, we carried out structure-based drug design on the substrate binding site of MTHFD2, by exploiting the cocrystal structure of MTHFD2 with the tricyclic coumarin-based inhibitor. The structure-based drug design campaign involves R-group enumeration, bioisostere replacement, molecular docking, ADME prediction, MM-GBSA binding free energy calculations, and molecular dynamics simulations, that led to a small library of new and potential compounds, capable of selectively inhibiting MTHFD2. The results reported herein are expected to benefit medicinal chemists working on the development of selective MTHFD2 inhibitors for cancer treatment, although experimental validation by biochemical and/or pharmacokinetic assays is required to substantiate the outcomes of the study.
Collapse
|
10
|
Scaletti ER, Gustafsson Westergren R, Andersson Y, Wiita E, Henriksson M, Homan EJ, Jemth A, Helleday T, Stenmark P. The First Structure of Human MTHFD2L and Its Implications for the Development of Isoform-Selective Inhibitors. ChemMedChem 2022; 17:e202200274. [PMID: 35712863 PMCID: PMC9796130 DOI: 10.1002/cmdc.202200274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/15/2022] [Indexed: 01/01/2023]
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a mitochondrial 1-carbon metabolism enzyme, which is an attractive anticancer drug target as it is highly upregulated in cancer but is not expressed in healthy adult cells. Selective MTHFD2 inhibitors could therefore offer reduced side-effects during treatment, which are common with antifolate drugs that target other 1C-metabolism enzymes. This task is challenging however, as MTHFD2 shares high sequence identity with the constitutively expressed isozymes cytosolic MTHFD1 and mitochondrial MTHFD2L. In fact, one of the most potent MTHFD2 inhibitors reported to date, TH7299, is actually more active against MTHFD1 and MTHFD2L. While structures of MTHFD2 and MTHFD1 exist, no MTHFD2L structures are available. We determined the first structure of MTHFD2L and its complex with TH7299, which reveals the structural basis for its highly potent MTHFD2L inhibition. Detailed analysis of the MTHFD2L structure presented here clearly highlights the challenges associated with developing truly isoform-selective MTHFD2 inhibitors.
Collapse
Affiliation(s)
- Emma R. Scaletti
- Department of Biochemistry and BiophysicsStockholm UniversitySvante Arrhenius väg 16 CStockholm106 91Sweden
| | | | - Yasmin Andersson
- Drug Discovery and Development Platform, Science for Life Laboratory School of BiotechnologyRoyal Institute of TechnologyTomtebodavägen 23aStockholm17165Sweden
| | - Elisee Wiita
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Martin Henriksson
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Evert J. Homan
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Ann‐Sofie Jemth
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Thomas Helleday
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
- Department of Oncology and MetabolismThe University of SheffieldBeech Hill RoadSheffieldS10 2RXUK
| | - Pål Stenmark
- Department of Biochemistry and BiophysicsStockholm UniversitySvante Arrhenius väg 16 CStockholm106 91Sweden
| |
Collapse
|
11
|
Huang S, Luo Q, Huang J, Wei J, Wang S, Hong C, Qiu P, Li C. A Cluster of Metabolic-Related Genes Serve as Potential Prognostic Biomarkers for Renal Cell Carcinoma. Front Genet 2022; 13:902064. [PMID: 35873461 PMCID: PMC9301649 DOI: 10.3389/fgene.2022.902064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/07/2022] [Indexed: 12/03/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of renal cancer, characterized by the dysregulation of metabolic pathways. RCC is the second highest cause of death among patients with urologic cancers and those with cancer cell metastases have a 5-years survival rate of only 10–15%. Thus, reliable prognostic biomarkers are essential tools to predict RCC patient outcomes. This study identified differentially expressed genes (DEGs) in the gene expression omnibus (GEO) database that are associated with pre-and post-metastases in clear cell renal cell carcinoma (ccRCC) patients and intersected these with metabolism-related genes in the Kyoto encyclopedia of genes and genomes (KEGG) database to identify metabolism-related DEGs (DEMGs). GOplot and ggplot packages for gene ontology (GO) and KEGG pathway enrichment analysis of DEMGs with log (foldchange) (logFC) were used to identify metabolic pathways associated with DEMG. Upregulated risk genes and downregulated protective genes among the DEMGs and seven independent metabolic genes, RRM2, MTHFD2, AGXT2, ALDH6A1, GLDC, HOGA1, and ETNK2, were found using univariate and multivariate Cox regression analysis, intersection, and Lasso-Cox regression analysis to establish a metabolic risk score signature (MRSS). Kaplan-Meier survival curve of Overall Survival (OS) showed that the low-risk group had a significantly better prognosis than the high-risk group in both the training cohort (p < 0.001; HR = 2.73, 95% CI = 1.97–3.79) and the validation cohort (p = 0.001; HR = 2.84, 95% CI = 1.50–5.38). The nomogram combined with multiple clinical information and MRSS was more effective at predicting patient outcomes than a single independent prognostic factor. The impact of metabolism on ccRCC was also assessed, and seven metabolism-related genes were established and validated as biomarkers to predict patient outcomes effectively.
Collapse
|
12
|
The catalytic mechanism of the mitochondrial methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2). PLoS Comput Biol 2022; 18:e1010140. [PMID: 35613161 PMCID: PMC9173628 DOI: 10.1371/journal.pcbi.1010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/07/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) is a new drug target that is expressed in cancer cells but not in normal adult cells, which provides an Achilles heel to selectively kill cancer cells. Despite the availability of crystal structures of MTHFD2 in the inhibitor- and cofactor-bound forms, key information is missing due to technical limitations, including (a) the location of absolutely required Mg2+ ion, and (b) the substrate-bound form of MTHFD2. Using computational modeling and simulations, we propose that two magnesium ions are present at the active site whereby (i) Arg233, Asp225, and two water molecules coordinate MgA2+, while MgA2+ together with Arg233 stabilize the inorganic phosphate (Pi); (ii) Asp168 and three water molecules coordinate MgB2+, and MgB2+ further stabilizes Pi by forming a hydrogen bond with two oxygens of Pi; (iii) Arg201 directly coordinates the Pi; and (iv) through three water-mediated interactions, Asp168 contributes to the positioning and stabilization of MgA2+, MgB2+ and Pi. Our computational study at the empirical valence bond level allowed us also to elucidate the detailed reaction mechanisms. We found that the dehydrogenase activity features a proton-coupled electron transfer with charge redistribution connected to the reorganization of the surrounding water molecules which further facilitates the subsequent cyclohydrolase activity. The cyclohydrolase activity then drives the hydration of the imidazoline ring and the ring opening in a concerted way. Furthermore, we have uncovered that two key residues, Ser197/Arg233, are important factors in determining the cofactor (NADP+/NAD+) preference of the dehydrogenase activity. Our work sheds new light on the structural and kinetic framework of MTHFD2, which will be helpful to design small molecule inhibitors that can be used for cancer treatment.
Collapse
|