1
|
Jakobsen S, Nielsen CU. Exploring Amino Acid Transporters as Therapeutic Targets for Cancer: An Examination of Inhibitor Structures, Selectivity Issues, and Discovery Approaches. Pharmaceutics 2024; 16:197. [PMID: 38399253 PMCID: PMC10893028 DOI: 10.3390/pharmaceutics16020197] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Amino acid transporters are abundant amongst the solute carrier family and have an important role in facilitating the transfer of amino acids across cell membranes. Because of their impact on cell nutrient distribution, they also appear to have an important role in the growth and development of cancer. Naturally, this has made amino acid transporters a novel target of interest for the development of new anticancer drugs. Many attempts have been made to develop inhibitors of amino acid transporters to slow down cancer cell growth, and some have even reached clinical trials. The purpose of this review is to help organize the available information on the efforts to discover amino acid transporter inhibitors by focusing on the amino acid transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), xCT (SLC7A11), SNAT1 (SLC38A1), SNAT2 (SLC38A2), and PAT1 (SLC36A1). We discuss the function of the transporters, their implication in cancer, their known inhibitors, issues regarding selective inhibitors, and the efforts and strategies of discovering inhibitors. The goal is to encourage researchers to continue the search and development within the field of cancer treatment research targeting amino acid transporters.
Collapse
Affiliation(s)
- Sebastian Jakobsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
2
|
Qin L, Cheng X, Wang S, Gong G, Su H, Huang H, Chen T, Damdinjav D, Dorjsuren B, Li Z, Qiu Z, Bian J. Discovery of Novel Aminobutanoic Acid-Based ASCT2 Inhibitors for the Treatment of Non-Small-Cell Lung Cancer. J Med Chem 2024; 67:988-1007. [PMID: 38217503 DOI: 10.1021/acs.jmedchem.3c01093] [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: 01/15/2024]
Abstract
Alanine-serine-cysteine transporter 2 (ASCT2) is up-regulated in lung cancers, and inhibiting it could potentially lead to nutrient deprivation, making it a viable strategy for cancer treatment. In this study, we present a series of ASCT2 inhibitors based on aminobutanoic acids, which exhibit potent inhibitory activity. Two compounds, 20k and 25e, were identified as novel and potent ASCT2 inhibitors, with IC50 values at the micromolar level in both A549 and HEK293 cells, effectively blocking glutamine (Gln) uptake. Additionally, these compounds regulated amino acid metabolism, suppressed mTOR signaling, inhibited non-small-cell lung cancer (NSCLC) growth, and induced apoptosis. In vivo, experiments showed that 20k and 25e suppressed tumor growth in an A549 xenograft model, with tumor growth inhibition (TGI) values of 65 and 70% at 25 mg/kg, respectively, while V9302 only achieved a TGI value of 29%. Furthermore, both compounds demonstrated promising therapeutic potential in patient-derived organoids. Therefore, these ASCT2 inhibitors based on aminobutanoic acids are promising therapeutic agents for treating NSCLC by targeting cancer Gln metabolism.
Collapse
Affiliation(s)
- Lian Qin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Xinying Cheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Shijiao Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Guangyue Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Huiyan Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Huidan Huang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, P. R. China
| | - Tian Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Science, Ulaanbaatar 14210, Mongolia
| | - Buyankhishig Dorjsuren
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Zhixia Qiu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211100, P. R. China
| | - Jinlei Bian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, P. R. China
| |
Collapse
|
3
|
Lyu XD, Liu Y, Wang J, Wei YC, Han Y, Li X, Zhang Q, Liu ZR, Li ZZ, Jiang JW, Hu HL, Yuan ST, Sun L. A Novel ASCT2 Inhibitor, C118P, Blocks Glutamine Transport and Exhibits Antitumour Efficacy in Breast Cancer. Cancers (Basel) 2023; 15:5082. [PMID: 37894450 PMCID: PMC10605716 DOI: 10.3390/cancers15205082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The microtubule protein inhibitor C118P shows excellent anti-breast cancer effects. However, the potential targets and mechanisms of C118P in breast cancer remain unknown. METHODS Real-time cellular analysis (RTCA) was used to detect cell viability. Apoptosis and the cell cycle were detected by flow cytometry. Computer docking simulations, surface plasmon resonance (SPR) technology, and microscale thermophoresis (MST) were conducted to study the interaction between C118P and alanine-serine-cysteine transporter 2 (ASCT2). Seahorse XF technology was used to measure the basal oxygen consumption rate (OCR). The effect of C118P in the adipose microenvironment was explored using a co-culture model of adipocytes and breast cancer cells and mouse cytokine chip. RESULTS C118P inhibited proliferation, potentiated apoptosis, and induced G2/M cell cycle arrest in breast cancer cells. Notably, ASCT2 was validated as a C118P target through reverse docking, SPR, and MST. C118P suppressed glutamine metabolism and mediated autophagy via ASCT2. Similar results were obtained in the adipocyte-breast cancer microenvironment. Adipose-derived interleukin-6 (IL-6) promoted the proliferation of breast cancer cells by enhancing glutamine metabolism via ASCT2. C118P inhibited the upregulation of ASCT2 by inhibiting the effect of IL-6 in co-cultures. CONCLUSION C118P exerts an antitumour effect against breast cancer via the glutamine transporter ASCT2.
Collapse
Affiliation(s)
- Xiao-Dan Lyu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Yang Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Yuan-Cheng Wei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Yi Han
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Xue Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Qian Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Zheng-Rui Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Zheng-Zheng Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| | - Jing-Wei Jiang
- Shuangyun BioMed Sci & Tech Co., Ltd., Suzhou 215000, China;
| | - Hao-Lin Hu
- General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Sheng-Tao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; (X.-D.L.); (Y.L.); (J.W.); (Y.-C.W.); (Y.H.); (X.L.); (Q.Z.); (Z.-R.L.); (Z.-Z.L.)
| |
Collapse
|
4
|
Pommereau A, Licher T, Bärenz F. Solid-Supported Membrane (SSM)-Based Electrophysiology Assays Using Surface Electrogenic Event Reader Technology (SURFE²R) in Early Drug Discovery. Curr Protoc 2023; 3:e650. [PMID: 36912603 DOI: 10.1002/cpz1.650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
This article presents detailed descriptions of procedures and troubleshooting tips for solid-supported membrane (SSM)-based electrophysiology assays (SURFE²R) to measure electrogenic solute carrier transporter proteins (SLCs) and assess the effects of compounds that modulate their activity. SURFE²R allows the use of the standard 96-well format, making it an ideal platform for tertiary assays in a drug-discovery campaign. The assays are performed with cell-line-derived membrane fractions or proteoliposomes containing the transporter of interest. Three main protocols are described for the isolation of membrane fractions from cell culture and the generation of proteoliposomes containing the transporter of interest. Additionally, detailed protocols for SURFE²R single concentration and dose-response experiments are included to measure the potencies of test compounds in stimulating or inhibiting transporter function (EC50 or IC50 values, respectively) and kinetic functional assays to calculate apparent affinity (kM ) and maximal velocity (Vmax ) of substrate uptake. © 2023 Sanofi. Current Protocols published by Wiley Periodicals LLC. PROTOCOL GROUP 1: Sample preparation for SSM-based electrophysiology assays Support Protocol 1: Production of cell batches Support Protocol 2: Simple isolation of cell membranes Alternate Protocol 1: Isolation of cell membranes with sucrose gradient pre-purification Support Protocol 3: Production and isolation of liposomes Support Protocol 4: Preparation of sensor with isolated cell membranes Alternate Protocol 2: Preparation of sensor with isolated proteoliposomes PROTOCOL GROUP 2: Determination of assay parameters for SSM-based electrophysiology assay Support Protocol 5: Assay with stable buffer Alternate Protocol 3: Assay with ion gradient Support Protocol 6: Determination of membrane/liposome concentration Support Protocol 7: Determination of substrate dependency kM PROTOCOL GROUP 3: Determination of advanced assay parameters for SSM-based electrophysiology assays Support Protocol 8: Assessment of ion concentration dependency Support Protocol 9: Assessment of pH dependency Support Protocol 10: Assessment of DMSO dependency Support Protocol 11: Assessment of signal stability with multiple activations PROTOCOL GROUP 4: Compound testing through SSM-based electrophysiology assays using SURFE²R apparatus Support Protocol 12: Assessment of signal specificity of a published inhibitor or unknown compound(s) Support Protocol 13: Compound wash-out Support Protocol 14: Statistical analysis.
Collapse
Affiliation(s)
- Antje Pommereau
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Germany
| | - Thomas Licher
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Germany
| | - Felix Bärenz
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Germany
| |
Collapse
|
5
|
Chang Y, Hawkins BA, Du JJ, Groundwater PW, Hibbs DE, Lai F. A Guide to In Silico Drug Design. Pharmaceutics 2022; 15:pharmaceutics15010049. [PMID: 36678678 PMCID: PMC9867171 DOI: 10.3390/pharmaceutics15010049] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
The drug discovery process is a rocky path that is full of challenges, with the result that very few candidates progress from hit compound to a commercially available product, often due to factors, such as poor binding affinity, off-target effects, or physicochemical properties, such as solubility or stability. This process is further complicated by high research and development costs and time requirements. It is thus important to optimise every step of the process in order to maximise the chances of success. As a result of the recent advancements in computer power and technology, computer-aided drug design (CADD) has become an integral part of modern drug discovery to guide and accelerate the process. In this review, we present an overview of the important CADD methods and applications, such as in silico structure prediction, refinement, modelling and target validation, that are commonly used in this area.
Collapse
Affiliation(s)
- Yiqun Chang
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Bryson A. Hawkins
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jonathan J. Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Paul W. Groundwater
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David E. Hibbs
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Felcia Lai
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
6
|
Ndaru E, Zielewicz L, Shi Y, Hutchinson K, Garibsingh RAA, Schlessinger A, Grewer C. Alanine serine cysteine transporter (ASCT) substrate binding site properties probed with hydroxyhomoserine esters. J PHYS ORG CHEM 2022; 35:e4347. [PMID: 36568026 PMCID: PMC9786560 DOI: 10.1002/poc.4347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
The glutamine transporter ASCT2 is highly overexpressed in cancer cells. Block of glutamine uptake by ASCT2 is a potential strategy to inhibit growth of cancer cells. However, pharmacology of the ASCT2 binding site is not well established. In this work, we report the computational docking to the binding site, and the synthesis of a new class of ASCT2 inhibitors based on the novel L-hydroxyhomoserine scaffold. While these compounds inhibit the ASCT2 leak anion conductance, as expected for competitive inhibitors, they did not block leak conductance in glutamate transporters (EAAT1-3 and EAAT5). They were also ineffective with respect to subtype ASCT1, which has >57% amino acid sequence similarity to ASCT2. Molecular docking studies agree very well with the experimental results and suggest specific polar interactions in the ASCT2 binding site. Our findings add to the repertoire of ASCT2 inhibitors and will aid in further studies of ASCT2 pharmacology.
Collapse
Affiliation(s)
- Elias Ndaru
- Department of Chemistry, Binghamton University, Binghamton, NY 13902
| | - Laura Zielewicz
- Department of Chemistry, Binghamton University, Binghamton, NY 13902
| | - Yueyue Shi
- Department of Chemistry, Binghamton University, Binghamton, NY 13902
| | - Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Rachel-Ann A Garibsingh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Christof Grewer
- Department of Chemistry, Binghamton University, Binghamton, NY 13902
| |
Collapse
|
7
|
Lan Q, Zhao Z, Liao H, Zheng F, Chen Y, Wu T, Tian Y, Pang J. Mutation in Transmembrane Domain 8 of Human Urate Transporter 1 Disrupts Uric Acid Recognition and Transport. ACS OMEGA 2022; 7:34621-34631. [PMID: 36188325 PMCID: PMC9521027 DOI: 10.1021/acsomega.2c04543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Human urate transporter 1 (hURAT1) is the most pivotal therapeutic target for hyperuricemia. Due to a lack of crystal structure information, the atomic structure of URAT1 is not clearly understood. In this study, a multiple sequence alignment was performed, and K393, a positively charged residue in transmembrane domain (TMD) 8, was observed to be highly conserved in organic anion transporters (OATs). K393 was substituted with a positively, negatively, and neutrally charged amino acid via site-directed mutagenesis and then used to transfect HEK293 cells. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) analyses indicated that mutants of K393 showed mRNA and protein expression levels similar to those in the WT group. The nonpositively charged mutants K393A, K393D, and K393E eliminated 70-80% of 14C-uric acid transport capacity, while the K393H mutant showed slight and the K393R mutant showed no reduced transport capacity compared with the WT group. Binding assays indicated that K393A, K393D, and K393E conferred lowered uric acid binding affinity. As indicated by the K m and V max values obtained from saturation kinetic experiments, K393A, K393D, and K393E showed increased K m values, but K393R and K393H showed K m values similar to those in the WT group. K393 also contributed to a high affinity for benzbromarone (BM) interaction. The inhibitory effects of BM were partly abolished in K393 mutants, with increased IC50 values compared with the WT group. BM also exhibited weaker inhibitory effects on 14C-uric acid binding in K393R and K393H mutants. In an outward homology model of URAT1, K393 was located in the inner part of the transport tunnel, and further molecular docking analysis indicated that uric acid and BM showed possible hydrogen bonds with K393. Mutants K393R and K393H showed possible interactions with uric acid, and positive charges confer high affinity for uric acid as revealed by their surface electrostatic potential. In conclusion, our data provide evidence that K393 is an important residue for the recognition of uric acid or inhibitors by URAT1.
Collapse
Affiliation(s)
- Qunsheng Lan
- Department
of Pharmacy, Shenzhen Longhua District Central
Hospital, No. 187 Western
Guanlan Avenue, Shenzhen, Guangdong 518110, China
| | - Ze’an Zhao
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hui Liao
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fengxin Zheng
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yongjun Chen
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ting Wu
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuanxin Tian
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianxin Pang
- Guangdong
Provincial Key Laboratory of Drug Screening, School of Pharmaceutical
Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
8
|
Duan Z, Zhou Z, Lu F, Zhang Y, Guo X, Gui C, Zhang H. Antitumor activity of mianserin (a tetracyclic antidepressant) primarily driven by the inhibition of SLC1A5-mediated glutamine transport. Invest New Drugs 2022; 40:977-989. [PMID: 35834041 DOI: 10.1007/s10637-022-01284-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
Abstract
Targeting tumor metabolic vulnerabilities such as "glutamine addiction" has become an attractive approach for the discovery of novel antitumor agents. Among various mechanisms explored, SLC1A5, a membrane transporter that plays an important role in glutamine cellular uptake, represents a viable target to interfere with tumor's ability to acquire critical nutrients during proliferation. In the present study, a stably transfected HEK293 cell line with human SLC1A5 (HEK293-SLC1A5) was established for the screening and identification of small molecule SLC1A5 inhibitors. This in vitro system, in conjunction with direct measurement of SLC1A5-mediated L-glutamine-2,3,3,4,4-D5 (substrate) uptake, was practical and efficient in ensuring the specificity of SLC1A5 inhibition. Among a group of diverse compounds tested, mianserin (a tetracyclic antidepressant) demonstrated a marked inhibition of SLC1A5-mediated glutamine uptake. Subsequent investigations using SW480 cells demonstrated that mianserin was capable of inhibiting SW480 tumor growth both in vitro and in vivo, and the in vivo antitumor efficacy was correlated to the reduction of glutamine concentrations in tumor tissues. Computational analysis revealed that hydrophobic interactions between SLC1A5 and its inhibitors could be a critical factor in drug design. Taken together, the current findings confirmed the feasibility of targeting SLC1A5-mediated glutamine uptake as a novel approach for antitumor intervention. It is anticipated that structural insights obtained based on homology modeling would lead to the discovery of more potent and specific SLC1A5 inhibitors for clinical development.
Collapse
Affiliation(s)
- Zelin Duan
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhiyun Zhou
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Feifei Lu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yawen Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xvqin Guo
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Chunshan Gui
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Hongjian Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
9
|
Rational design of ASCT2 inhibitors using an integrated experimental-computational approach. Proc Natl Acad Sci U S A 2021; 118:2104093118. [PMID: 34507995 PMCID: PMC8449414 DOI: 10.1073/pnas.2104093118] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 12/04/2022] Open
Abstract
The glutamine transporter ASCT2 is an emerging therapeutic target for various cancer types. Here, we use an integrated computational and experimental approach to develop unique ASCT2 inhibitors targeting a conformational state useful for rational drug design. We apply computational chemistry tools such as molecular docking and molecular dynamics simulations, in combination with structure determination with cryo-electron microscopy and synthetic chemistry, to design multiple ASCT2 inhibitors. Our results reveal a unique mechanism of stereospecific inhibition of ASCT2 and highlight the utility of combining state-of-the-art computational and experimental approaches in characterizing challenging human membrane protein targets. ASCT2 (SLC1A5) is a sodium-dependent neutral amino acid transporter that controls amino acid homeostasis in peripheral tissues. In cancer, ASCT2 is up-regulated where it modulates intracellular glutamine levels, fueling cell proliferation. Nutrient deprivation via ASCT2 inhibition provides a potential strategy for cancer therapy. Here, we rationally designed stereospecific inhibitors exploiting specific subpockets in the substrate binding site using computational modeling and cryo-electron microscopy (cryo-EM). The final structures combined with molecular dynamics simulations reveal multiple pharmacologically relevant conformations in the ASCT2 binding site as well as a previously unknown mechanism of stereospecific inhibition. Furthermore, this integrated analysis guided the design of a series of unique ASCT2 inhibitors. Our results provide a framework for future development of cancer therapeutics targeting nutrient transport via ASCT2, as well as demonstrate the utility of combining computational modeling and cryo-EM for solute carrier ligand discovery.
Collapse
|
10
|
Gulezian E, Crivello C, Bednenko J, Zafra C, Zhang Y, Colussi P, Hussain S. Membrane protein production and formulation for drug discovery. Trends Pharmacol Sci 2021; 42:657-674. [PMID: 34270922 DOI: 10.1016/j.tips.2021.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Integral membrane proteins (MPs) are important drug targets across most fields of medicine, but historically have posed a major challenge for drug discovery due to difficulties in producing them in functional forms. We review the state of the art in drug discovery strategies using recombinant multipass MPs, and outline methods to successfully express, stabilize, and formulate them for small-molecule and monoclonal antibody therapeutics development. Advances in structure-based drug design and high-throughput screening are allowing access to previously intractable targets such as ion channels and transporters, propelling the field towards the development of highly specific therapies targeting desired conformations.
Collapse
Affiliation(s)
- Ellen Gulezian
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | | | - Janna Bednenko
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Claudia Zafra
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Yihui Zhang
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Paul Colussi
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA
| | - Sunyia Hussain
- TetraGenetics Inc., 91 Mystic Street, Arlington, MA 02474, USA.
| |
Collapse
|
11
|
Dvorak V, Wiedmer T, Ingles-Prieto A, Altermatt P, Batoulis H, Bärenz F, Bender E, Digles D, Dürrenberger F, Heitman LH, IJzerman AP, Kell DB, Kickinger S, Körzö D, Leippe P, Licher T, Manolova V, Rizzetto R, Sassone F, Scarabottolo L, Schlessinger A, Schneider V, Sijben HJ, Steck AL, Sundström H, Tremolada S, Wilhelm M, Wright Muelas M, Zindel D, Steppan CM, Superti-Furga G. An Overview of Cell-Based Assay Platforms for the Solute Carrier Family of Transporters. Front Pharmacol 2021; 12:722889. [PMID: 34447313 PMCID: PMC8383457 DOI: 10.3389/fphar.2021.722889] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
The solute carrier (SLC) superfamily represents the biggest family of transporters with important roles in health and disease. Despite being attractive and druggable targets, the majority of SLCs remains understudied. One major hurdle in research on SLCs is the lack of tools, such as cell-based assays to investigate their biological role and for drug discovery. Another challenge is the disperse and anecdotal information on assay strategies that are suitable for SLCs. This review provides a comprehensive overview of state-of-the-art cellular assay technologies for SLC research and discusses relevant SLC characteristics enabling the choice of an optimal assay technology. The Innovative Medicines Initiative consortium RESOLUTE intends to accelerate research on SLCs by providing the scientific community with high-quality reagents, assay technologies and data sets, and to ultimately unlock SLCs for drug discovery.
Collapse
Affiliation(s)
- Vojtech Dvorak
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Helena Batoulis
- Drug Discovery Sciences–Lead Discovery, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Felix Bärenz
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Eckhard Bender
- Drug Discovery Sciences–Lead Discovery, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Daniela Digles
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | | | - Laura H. Heitman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Stefanie Kickinger
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Daniel Körzö
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Philipp Leippe
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Thomas Licher
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | | | | | | | | | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Vanessa Schneider
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Hubert J. Sijben
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | | | | | | | | | - Marina Wright Muelas
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Diana Zindel
- Drug Discovery Sciences–Lead Discovery, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Claire M. Steppan
- Pfizer Worldwide Research, Development and Medical, Groton, MA, United States
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Recent developments in ligands and chemical probes targeting solute carrier transporters. Curr Opin Chem Biol 2021; 62:53-63. [PMID: 33689964 DOI: 10.1016/j.cbpa.2021.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/12/2021] [Accepted: 01/31/2021] [Indexed: 12/30/2022]
Abstract
Solute carrier (SLC) membrane transporters remain a largely unexploited target class, despite their central roles in cell identity and metabolism. This gap is reflected in the lack of high-quality chemical ligands or probes and in the small number of compounds that have progressed toward clinical development. In this review, we discuss recent advancements in SLC ligand discovery as well as new candidates that have been added to the investigational toolkit, with a particular focus on first-in-class ligands and the cognate discovery strategies. The availability of new probes expands the opportunity to elucidate the functions of SLCs and their relevance in physiology and explores any future potential of SLC druggability.
Collapse
|
13
|
Zhao Z, Jiang Y, Li L, Chen Y, Li Y, Lan Q, Wu T, Lin C, Cao Y, Nandakumar KS, Zhou P, Tian Y, Pang J. Structural Insights into the Atomistic Mechanisms of Uric Acid Recognition and Translocation of Human Urate Anion Transporter 1. ACS OMEGA 2020; 5:33421-33432. [PMID: 33403304 PMCID: PMC7774290 DOI: 10.1021/acsomega.0c05360] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Background: Human urate transporter 1 (hURAT1) is the most pivotal therapeutic target for treating hyperuricemia. However, the molecular interactions between uric acid and URAT1 are still unknown due to lack of structural details. Methods: In the present study, several methods (homology modeling, sequence alignment, docking, and mutagenesis) were used to explain the atomistic mechanisms of uric acid transport of hURAT1. Results: Residues W357-F365 in the TMD7 and P484-R487 in the TMD11 present in the hURAT1 have unique roles in both binding to the uric acid and causing subsequent structural changes. These residues, located in the transport tunnel, were found to be related to the structural changes, as demonstrated by the reduced V max values and an unaltered expression of protein level. In addition, W357, G361, T363, F365, and R487 residues may confer high affinity for binding to uric acid. An outward-open homology model of hURAT1 revealed a crucial role for these two domains in the conformational changes of hURAT1. F241 and H245 in TMD5, and R477 and R487 in TMD11 may confer high affinity for uric acid, and as the docking analysis suggests, they may also enhance the affinity for the inhibitors. R477 relation to the structural changes was demonstrated by the V max values of the mutants and the contribution of positive charge to the uric acid selectivity. Conclusions: W357-F365 in TMD7, P484-R487 in TMD11, and residues F241, H245, and R477 were found to be critical for the translocation and recognition of uric acid.
Collapse
|
14
|
Interaction of the neutral amino acid transporter ASCT2 with basic amino acids. Biochem J 2020; 477:1443-1457. [PMID: 32242892 DOI: 10.1042/bcj20190859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/16/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
Abstract
Glutamine transport across cell membranes is performed by a variety of transporters, including the alanine serine cysteine transporter 2 (ASCT2). The substrate-binding site of ASCT2 was proposed to be specific for small amino acids with neutral side chains, excluding basic substrates such as lysine. A series of competitive inhibitors of ASCT2 with low µM affinity were developed previously, on the basis of the 2,4-diaminobutyric acid (DAB) scaffold with a potential positive charge in the side chain. Therefore, we tested whether basic amino acids with side chains shorter than lysine can interact with the ASCT2 binding site. Molecular docking of L-1,3-diaminopropionic acid (L-DAP) and L-DAB suggested that these compounds bind to ASCT2. Consistent with this prediction, L-DAP and L-DAB, but not ornithine, lysine or D-DAP, elicited currents when applied to ASCT2-expressing cells. The currents were carried by anions and showed the hallmark properties of ASCT2 currents induced by transported substrates. The L-DAP response could be eliminated by a competitive ASCT2 inhibitor, suggesting that binding occurs at the substrate binding site. The KM for L-DAP was weakly voltage dependent. Furthermore, the pH dependence of the L-DAP response showed that the compound can bind in several protonation states. Together, these results suggest that the ASCT2 binding site is able to recognize L-amino acids with short, basic side chains, such as the L-DAP derivative β-N-methylamino-l-Alanine (BMAA), a well-studied neurotoxin. Our results expand the substrate specificity of ASCT2 to include amino acid substrates with positively charged side chains.
Collapse
|
15
|
Bröer S. Amino Acid Transporters as Targets for Cancer Therapy: Why, Where, When, and How. Int J Mol Sci 2020; 21:ijms21176156. [PMID: 32859034 PMCID: PMC7503255 DOI: 10.3390/ijms21176156] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/23/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Amino acids are indispensable for the growth of cancer cells. This includes essential amino acids, the carbon skeleton of which cannot be synthesized, and conditionally essential amino acids, for which the metabolic demands exceed the capacity to synthesize them. Moreover, amino acids are important signaling molecules regulating metabolic pathways, protein translation, autophagy, defense against reactive oxygen species, and many other functions. Blocking uptake of amino acids into cancer cells is therefore a viable strategy to reduce growth. A number of studies have used genome-wide silencing or knock-out approaches, which cover all known amino acid transporters in a large variety of cancer cell lines. In this review, these studies are interrogated together with other databases to identify vulnerabilities with regard to amino acid transport. Several themes emerge, such as synthetic lethality, reduced redundancy, and selective vulnerability, which can be exploited to stop cancer cell growth.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Canberra ACT 2600, Australia
| |
Collapse
|
16
|
Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine transporters as pharmacological targets: From function to drug design. Asian J Pharm Sci 2020; 15:207-219. [PMID: 32373200 PMCID: PMC7193454 DOI: 10.1016/j.ajps.2020.02.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/18/2020] [Accepted: 02/29/2020] [Indexed: 12/17/2022] Open
Abstract
Among the different targets of administered drugs, there are membrane transporters that play also a role in drug delivery and disposition. Moreover, drug-transporter interactions are responsible for off-target effects of drugs underlying their toxicity. The improvement of the drug design process is subjected to the identification of those membrane transporters mostly relevant for drug absorption, delivery and side effect production. A peculiar group of proteins with great relevance to pharmacology is constituted by the membrane transporters responsible for managing glutamine traffic in different body districts. The interest around glutamine metabolism lies in its physio-pathological role; glutamine is considered a conditionally essential amino acid because highly proliferative cells have an increased request of glutamine that cannot be satisfied only by endogenous synthesis. Then, glutamine transporters provide cells with this special nutrient. Among the glutamine transporters, SLC1A5, SLC6A14, SLC6A19, SLC7A5, SLC7A8 and some members of SLC38 family are the best characterized, so far, in both physiological and pathological conditions. Few 3D structures have been solved by CryoEM; other structural data on these transporters have been obtained by computational analysis. Interactions with drugs have been described for several transporters of this group. For some of them, the studies are at an advanced stage, for others, the studies are still in nuce and novel biochemical findings open intriguing perspectives.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lorena Pochini
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lara Console
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| |
Collapse
|
17
|
Zhang L, Sui C, Yang W, Luo Q. Amino acid transporters: Emerging roles in drug delivery for tumor-targeting therapy. Asian J Pharm Sci 2020; 15:192-206. [PMID: 32373199 PMCID: PMC7193455 DOI: 10.1016/j.ajps.2019.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/22/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
Amino acid transporters, which play a vital role in transporting amino acids for the biosynthesis of mammalian cells, are highly expressed in types of tumors. Increasing studies have shown the feasibility of amino acid transporters as a component of tumor-targeting therapy. In this review, we focus on tumor-related amino acid transporters and their potential use in tumor-targeting therapy. Firstly, the expression characteristics of amino acid transporters in cancer and their relationship with tumor growth are reviewed. Secondly, the recognition requirements are discussed, focusing on the "acid-base" properties, conformational isomerism and structural analogues. Finally, recent developments in amino acid transporter-targeting drug delivery strategies are highlighted, including prodrugs and nanocarriers, with special attention to the latest findings of molecular mechanisms and targeting efficiency of transporter-mediated endocytosis. We aim to offer related clues that might lead to valuable tumor-targeting strategies by the utilization of amino acid transporters.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Chengguang Sui
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wenhan Yang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| |
Collapse
|
18
|
Amino Acid Transporters and Exchangers from the SLC1A Family: Structure, Mechanism and Roles in Physiology and Cancer. Neurochem Res 2020; 45:1268-1286. [DOI: 10.1007/s11064-019-02934-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
19
|
Garibsingh RAA, Schlessinger A. Advances and Challenges in Rational Drug Design for SLCs. Trends Pharmacol Sci 2019; 40:790-800. [PMID: 31519459 DOI: 10.1016/j.tips.2019.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 01/25/2023]
Abstract
There are over 420 human solute carrier (SLC) transporters from 65 families that are expressed ubiquitously in the body. The SLCs mediate the movement of ions, drugs, and metabolites across membranes and their dysfunction has been associated with a variety of diseases, such as diabetes, cancer, and central nervous system (CNS) disorders. Thus, SLCs are emerging as important targets for therapeutic intervention. Recent technological advances in experimental and computational biology allow better characterization of SLC pharmacology. Here we describe recent approaches to modulate SLC transporter function, with an emphasis on the use of computational approaches and computer-aided drug design (CADD) to study nutrient transporters. Finally, we discuss future perspectives in the rational design of SLC drugs.
Collapse
Affiliation(s)
- Rachel-Ann A Garibsingh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
20
|
Ndaru E, Garibsingh RAA, Shi Y, Wallace E, Zakrepine P, Wang J, Schlessinger A, Grewer C. Novel alanine serine cysteine transporter 2 (ASCT2) inhibitors based on sulfonamide and sulfonic acid ester scaffolds. J Gen Physiol 2019; 151:357-368. [PMID: 30718375 PMCID: PMC6400523 DOI: 10.1085/jgp.201812276] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 01/11/2023] Open
Abstract
The neutral amino acid transporter alanine serine cysteine transporter 2 (ASCT2) belongs to the solute carrier 1 (SLC1) family of transport proteins and transports neutral amino acids, such as alanine and glutamine, into the cell in exchange with intracellular amino acids. This amino acid transport is sodium dependent, but not driven by the transmembrane Na+ concentration gradient. Glutamine transport by ASCT2 is proposed to be important for glutamine homoeostasis in rapidly growing cancer cells to fulfill the energy and nitrogen demands of these cells. Thus, ASCT2 is thought to be a potential anticancer drug target. However, the pharmacology of the amino acid binding site is not well established. Here, we report on the synthesis and characterization of a novel class of ASCT2 inhibitors based on an amino acid scaffold with a sulfonamide/sulfonic acid ester linker to a hydrophobic group. The compounds were designed based on an improved ASCT2 homology model using the human glutamate transporter hEAAT1 crystal structure as a modeling template. The compounds were shown to inhibit with a competitive mechanism and a potency that scales with the hydrophobicity of the side chain. The most potent compound binds with an apparent affinity, K i, of 8 ± 4 µM and can block the alanine response with a K i of 40 ± 23 µM at 200 µM alanine concentration. Computational analysis predicts inhibitor interactions with the binding site through molecular docking. In conclusion, the sulfonamide/sulfonic acid ester scaffold provides facile synthetic access to ASCT2 inhibitors with a potentially large variability in chemical space of the hydrophobic side chain. These inhibitors will be useful chemical tools to further characterize the role of ASCT2 in disease as well as improve our understanding of inhibition mechanisms of this transporter.
Collapse
Affiliation(s)
- Elias Ndaru
- Department of Chemistry, Binghamton University, Binghamton, NY
| | - Rachel-Ann A Garibsingh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - YueYue Shi
- Department of Chemistry, Binghamton University, Binghamton, NY
| | - Evan Wallace
- Department of Chemistry, Binghamton University, Binghamton, NY
| | - Paul Zakrepine
- Department of Chemistry, Binghamton University, Binghamton, NY
| | - Jiali Wang
- Department of Chemistry, Binghamton University, Binghamton, NY
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Christof Grewer
- Department of Chemistry, Binghamton University, Binghamton, NY
| |
Collapse
|
21
|
Singh N, Scalise M, Galluccio M, Wieder M, Seidel T, Langer T, Indiveri C, Ecker GF. Discovery of Potent Inhibitors for the Large Neutral Amino Acid Transporter 1 (LAT1) by Structure-Based Methods. Int J Mol Sci 2018; 20:ijms20010027. [PMID: 30577601 PMCID: PMC6337383 DOI: 10.3390/ijms20010027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/15/2018] [Indexed: 12/20/2022] Open
Abstract
The large neutral amino acid transporter 1 (LAT1) is a promising anticancer target that is required for the cellular uptake of essential amino acids that serve as building blocks for cancer growth and proliferation. Here, we report a structure-based approach to identify chemically diverse and potent inhibitors of LAT1. First, a homology model of LAT1 that is based on the atomic structures of the prokaryotic homologs was constructed. Molecular docking of nitrogen mustards (NMs) with a wide range of affinity allowed for deriving a common binding mode that could explain the structure−activity relationship pattern in NMs. Subsequently, validated binding hypotheses were subjected to molecular dynamics simulation, which allowed for extracting a set of dynamic pharmacophores. Finally, a library of ~1.1 million molecules was virtually screened against these pharmacophores, followed by docking. Biological testing of the 30 top-ranked hits revealed 13 actives, with the best compound showing an IC50 value in the sub-μM range.
Collapse
Affiliation(s)
- Natesh Singh
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Mariafrancesca Scalise
- Department DiBEST, Unit of Biochemistry & Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy.
| | - Michele Galluccio
- Department DiBEST, Unit of Biochemistry & Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy.
| | - Marcus Wieder
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Thomas Seidel
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Thierry Langer
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Cesare Indiveri
- Department DiBEST, Unit of Biochemistry & Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy.
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| |
Collapse
|