1
|
Jiang R, Rao J, Su H, Li M. Degradable Metal-Small Molecule Drug Coordination Nanomedicines for DNA Damage-Augmented Cancer Chemodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501721. [PMID: 40331532 DOI: 10.1002/smll.202501721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/11/2025] [Indexed: 05/08/2025]
Abstract
The intrinsic DNA damage repair response represents a major limiting factor for the therapeutic efficacy of many anticancer therapies such as chemodynamic therapy (CDT). Here, a degradable metal-small molecule drug coordination nanomedicine (siAPE1/Cu-MP@HA) is developed for DNA damage-augmented CDT treatment of lung cancers. The design is achieved by the coordination-driven self-assembly of Cu2+ and 6-mercaptopurine (6-MP) with the siAPE1 loading and hyaluronic acid (HA) modification. The siAPE1/Cu-MP@HA possesses the tumor targeting ability and is degradable in lysosomes for specific releases of Cu2+, siAPE1, and 6-MP because of the weak acidity/glutathione (GSH) dual-responsiveness. Upon the depletion of endogenous GSH, the siAPE1/Cu-MP@HA promotes the ∙OH generation from endogenous H2O2 via the Fenton-like reaction, significantly enhancing the CDT anticancer effect. It is evidenced that the siAPE1/Cu-MP@HA treatment effectively disrupts the DNA synthesis and inhibits the DNA damage repair, thereby augmenting the DNA damage. In vivo, experiments demonstrate the remarkable therapeutic efficacy against A549 lung cancer in mice with negligible systemic toxicity after intravenous administration of the siAPE1/Cu-MP@HA. This study offers an innovative approach targeting the DNA damage repair system for enhancing CDT effects in cancer treatment.
Collapse
Affiliation(s)
- Renting Jiang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Jinchan Rao
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Hua Su
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| |
Collapse
|
2
|
Sołdatowska A, Urbanowicz M, Urbanowicz M, Sadowska K, Pijanowska DG. Exploring DNA-6MP interactions to develop a receptor with selective binding properties. Int J Biol Macromol 2025; 305:140910. [PMID: 39938824 DOI: 10.1016/j.ijbiomac.2025.140910] [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: 06/28/2024] [Revised: 01/03/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
This study focuses on identifying DNA sequences capable of selectively binding 6-mercaptopurine (6MP) or its analog, azathioprine (Aza), as a potential bioreceptor for biosensing methods. The approach used in the study is based on instrumental analysis methods such as UV-Vis spectroscopy, high performance liquid chromatography (HPLC), and nuclear magnetic resonance techniques to describe the interaction, its mode, and strength as well as selectivity of selected DNA sequences toward thiopurines. Thus, of the sequences tested, dsDNA GGCAGGACGGAG poses the ability to form complexes with 6MP, with an affinity constant of 2.52·103 M-1 as determined by DOSY NMR. Studies performed by HPLC technique confirmed the selectivity of the indicated strand toward selected interferents like natural metabolites and active pharmaceutical ingredients (API) used during azathioprine treatment. Further insights into the DNA-6MP interaction were provided by TOCSY and NOESY NMR experiments, highlighting the preference for guanine-rich sequences in binding, indicating a potential 6MP binding site in the dsDNA. The results also indicate the rapid conversion of azathioprine to 6-mercaptopurine upon contact with DNA, indirectly confirming the pro-drug nature of azathioprine. This research contributes to the development of DNA-based bioreceptors for selective determination of 6-mercaptopurine, with potential implications in personalized medicine and therapeutic drug monitoring.
Collapse
Affiliation(s)
- Anna Sołdatowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland.
| | - Marcin Urbanowicz
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland
| | - Magdalena Urbanowicz
- Falsified Medicines and Medical Materials Department, National Medicines Institute, Chełmska 30/34 St., 00-725 Warsaw, Poland
| | - Kamila Sadowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland
| | - Dorota G Pijanowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland
| |
Collapse
|
3
|
Wu AS, Mozessohn L, Kim RB, Zipursky JS. Severe myelosuppression and alopecia after thiopurine initiation in a patient with NUDT15 deficiency. Br J Clin Pharmacol 2025; 91:1511-1515. [PMID: 40099566 PMCID: PMC12035587 DOI: 10.1002/bcp.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Thiopurines are a class of immunosuppressant and antineoplastic agents. They are widely used in the treatment of inflammatory bowel disease, haematological malignancies and autoimmune diseases, but can cause significant toxicity. Inherited gene mutations are now recognized as independent risk factors for severe adverse drug reactions to thiopurines even at 10-fold dose reductions. We present a case of thiopurine toxicity resulting in severe myelosuppression, hepatotoxicity and alopecia in an individual with homozygous *3/*3 loss-of-function alleles in the NUDT15 gene. Our case highlights important differences in gene mutation frequencies between races that can help guide pharmacogenomic testing.
Collapse
Affiliation(s)
- Annie Siyu Wu
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Lee Mozessohn
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Division of Medical Oncology/HematologySunnybrook Health Sciences CentreTorontoOntarioCanada
- Odette Cancer CenterSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Richard B. Kim
- Department of MedicineUniversity of Western OntarioLondonOntarioCanada
- Division of Clinical Pharmacology and ToxicologyLondon Health Sciences CenterLondonOntarioCanada
| | - Jonathan S. Zipursky
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Division of General Internal MedicineSunnybrook Health Sciences CentreTorontoOntarioCanada
- Division of Clinical Pharmacology and ToxicologySunnybrook Health Sciences CentreTorontoOntarioCanada
- Sunnybrook Research InstituteTorontoOntarioCanada
| |
Collapse
|
4
|
Masson CD, Findlay-Greene F, Sousa FH, Henderson P, Fraser JA, Barlow PG, Stevens C. Characterisation of autophagy induction by the thiopurine drugs azathioprine, mercaptopurine and thioguanine in THP-1 macrophages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4467-4478. [PMID: 39485532 PMCID: PMC11978722 DOI: 10.1007/s00210-024-03563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Activating autophagy may be therapeutically beneficial, and we have previously shown that azathioprine (AZA), an immunomodulatory drug, induces autophagy. Here, we evaluated the induction of autophagy by the thiopurines AZA, mercaptopurine (6-MP) and thioguanine (6-TG) in THP-1 macrophages and investigated the mechanism of action in the context of this cellular process. The cytotoxicity of thiopurines was evaluated using an LDH assay. Induction of endogenous LC3 by thiopurines was evaluated using immunostaining. To confirm autophagy activation by thiopurines, a GFP-RFP-LC3 reporter plasmid was used to monitor the maturation of autophagosomes to autolysosomes. Induction of apoptosis by thiopurines was evaluated using Annexin V/PI staining, and ER stress was assessed via RT‒PCR analysis of XBP1 splicing. To gain insight into the mechanism of action of thiopurines, mTORC1 activity and eIF2α-S51 phosphorylation were evaluated by immunoblotting. Thiopurines were not cytotoxic to cells and induced strong time- and concentration-dependent autophagy. Thiopurines activate autophagy with complete progression through the pathway. Induction of autophagy by thiopurines occurred independently of apoptosis and ER stress. Immunoblotting revealed that AZA inhibited mTORC1 activity, and AZA and 6-TG increased eIF2α-S51 phosphorylation. In contrast, 6-MP had a minor effect on either signalling pathway. Thiopurines are strong inducers of autophagy, and autophagy induction should be considered among the mechanisms responsible for patient response to thiopurines.
Collapse
Affiliation(s)
- Connan D Masson
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Fern Findlay-Greene
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Filipa Henderson Sousa
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Paul Henderson
- Child Life and Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, EH16 4TJ, UK
| | - Jennifer A Fraser
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Peter G Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK.
| |
Collapse
|
5
|
Maillard M, Nguyen JQ, Yang W, Hoshitsuki K, Relling MV, Caudle KE, Crews KR, Jeha S, Inaba H, Pui CH, Bhatia S, Karol SE, Antillon-Klussmann FG, Haidar CE, Bhojwani D, Yang JJ. Clinical Actionability of the NUDT15 *4 (p.R139H) Allele and Its Association With Hispanic Ethnicity. Clin Pharmacol Ther 2025; 117:724-731. [PMID: 39688234 PMCID: PMC11995662 DOI: 10.1002/cpt.3501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/10/2024] [Indexed: 12/18/2024]
Abstract
Nudix hydrolase 15 (NUDT15) deficiency is strongly associated with thiopurine-induced myelosuppression. Currently, testing for NUDT15 deficiency is based on the genotyping of the most frequent and clinically characterized no-function variants, that is, *2, *3 and *9. The Hispanic/Latino-predominant variant NUDT15 *4 (p.R139H) is classified as "uncertain function" by the Clinical Pharmacogenetics Implementation Consortium, because of insufficient data to ascertain its clinical actionability. In this study, we evaluated the association of NUDT15 *4 with mercaptopurine (6-MP) tolerance in a retrospective cohort of 1,399 patients with acute lymphoblastic leukemia (ALL) of diverse ancestries. All patients were wildtype for thiopurine methyltransferase gene. Patients were treated with 6-MP in the context of ALL frontline clinical trials. The tolerated dose of 6-MP was used to assess drug toxicity during the maintenance phase of ALL therapy. Patients with NUDT15 *1/*4 (n = 16, all of whom self-identified as Hispanic/Latino) tolerated a significantly lower dose of 6-MP than did those with NUDT15 *1/*1: median [interquartile range] of 39.0 [21.2-52.8] mg/m2, vs. 62.2 [47.9-71.6] mg/m2, P value < 0.001. No patient homozygous for *4 was detected. In a separate retrospective validation study, six patients were identified as having NUDT15 *1/*4 by routine clinical pharmacogenetics testing and tolerated a 6-MP median dose of 38.7 mg/m2 (IQR, 33.7-54.0), confirming the need for dose reduction attributed to the NUDT15 *4 variant. These results point to the need to include NUDT15 *4 in pharmacogenetics-guided thiopurine dosing algorithms.
Collapse
Affiliation(s)
- Maud Maillard
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jenny Q. Nguyen
- Personalized Care Program, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Keito Hoshitsuki
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Mary V. Relling
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kelly E. Caudle
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kristine R. Crews
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sima Jeha
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship and Division of Pediatric Hematology-Oncology, University of Alabama at Birmingham, Alabama, USA
| | - Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Cyrine E. Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jun J. Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
6
|
Sanford SL, Badstübner M, Gerber M, Mannherz W, Lampl N, Dannenberg R, Hinchie A, Schaich MA, Myong S, Hedglin M, Agarwal S, Alder JK, Stone MD, Opresko PL. Chemotherapeutic 6-thio-2'-deoxyguanosine selectively targets and inhibits telomerase by inducing a non-productive telomere-bound telomerase complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636339. [PMID: 39975053 PMCID: PMC11838547 DOI: 10.1101/2025.02.05.636339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Most cancers upregulate the telomere lengthening enzyme telomerase to achieve unlimited cell division. How chemotherapeutic nucleoside 6-thio-2'-deoxyguanosine (6-thio-dG) targets telomerase to inhibit telomere maintenance in cancer cells and tumors was unclear. Here, we demonstrate that telomerase insertion of 6-thio-dGTP prevents synthesis of additional telomeric repeats but does not disrupt telomerase binding to telomeres. Specifically, 6-thio-dG inhibits telomere extension after telomerase translocates along its product DNA to reposition the template, inducing a non-productive complex rather than enzyme dissociation. Furthermore, we provide direct evidence that 6-thio-dG treatment inhibits telomere synthesis by telomerase in cancer cells. In agreement, telomerase-expressing cancer cells harboring critically short telomeres are more sensitive to 6-thio-dG and show a greater induction of telomere losses compared to cancer cells with long telomere reserves. Our studies reveal that telomere length and telomerase status determine 6-thio-dG sensitivity and uncover the molecular mechanism by which 6-thio-dG selectively inhibits telomerase synthesis of telomeric DNA.
Collapse
Affiliation(s)
| | - Mareike Badstübner
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - William Mannherz
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Noah Lampl
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Dannenberg
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Angela Hinchie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew A Schaich
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sua Myong
- Program in Cell, Molecular, Developmental Biology and Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Hedglin
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Suneet Agarwal
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jonathan K Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D. Stone
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Patricia L Opresko
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
7
|
Xu Z, Song Y, Sun J. Simultaneous production of singlet oxygen and superoxide anion by thiocarbonyl coumarin for photodynamic therapy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 327:125327. [PMID: 39500202 DOI: 10.1016/j.saa.2024.125327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 12/08/2024]
Abstract
Photodynamic therapy (PDT) is a medical treatment that kills target cells through reactive oxygen species (ROS) generated by photosensitizers (PS) and surrounding oxygen under the stimulus of light. Despite of its popularity in cancer treatment, PDT relys on oxygen and therefore suffers from long response time and low efficiency under low-oxygen situations such as tumor hypoxia. Herein, to improve the usage of oxygen and increase ROS yield, we synthesized six potential PSs termed DC-O, DC-S, DC-BrO, DC-BrS, DC-IO, and DC-IS, by modifying coumarins with thiocarbonyl and bromine/iodine. We found that the thiocarbonyl group induces a significant bathochromic shift of the absorption spectra. In addition, the ROS production was significantly improved, likely because these PSs can simultaneously generate singlet oxygen (1O2) and superoxide anions (O2•-) through different pathways. Among these compounds, DC-BrS produces largest amount of ROS and exhibits strongest cytotoxicity towards cells, the survival rate of B16-F10 cells incubated with DC-BrS was only 20.7 % after irradiation at 460 nm for 10 min, indicating DC-BrS as a strong candidate for photodynamic therapy. Most importantly, this work provides an important direction for the design of PSs in the future.
Collapse
Affiliation(s)
- Zhijing Xu
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Yingzhuang Song
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Jinyu Sun
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 Gaoxin Nanjiu Road, Shenzhen, Guangdong 518057, China; Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
8
|
Baek S, Hong S, Kim E, Park S, Lee M, Park J, Cho Y, Yoon H, Kim D, Yun Y, Kim Y, Choi Y, Kang K, Jung S, Kim JP, Kim E, Seo SW, Jung Y, Jo D. A Novel RAGE Modulator Induces Soluble RAGE to Reduce BACE1 Expression in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407812. [PMID: 39755927 PMCID: PMC11848596 DOI: 10.1002/advs.202407812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/12/2024] [Indexed: 01/06/2025]
Abstract
β-secretase (BACE1) is instrumental in amyloid-β (Aβ) production, with overexpression noted in Alzheimer's disease (AD) neuropathology. The interaction of Aβ with the receptor for advanced glycation endproducts (RAGE) facilitates cerebral uptake of Aβ and exacerbates its neurotoxicity and neuroinflammation, further augmenting BACE1 expression. Given the limitations of previous BACE1 inhibition efforts, the study explores reducing BACE1 expression to mitigate AD pathology. The research reveals that the anticancer agent 6-thioguanosine (6-TG) markedly diminishes BACE1 expression without eliciting cytotoxicity while enhancing microglial phagocytic activity, and ameliorate cognitive impairments with reducing Aβ accumulation in AD mice. Leveraging advanced deep learning-based tool for target identification, and corroborating with surface plasmon resonance assays, it is elucidated that 6-TG directly interacts with RAGE, modulating BACE1 expression through the JAK2-STAT1 pathway and elevating soluble RAGE (sRAGE) levels in the brain. The findings illuminate the therapeutic potential of 6-TG in ameliorating AD manifestations and advocate for small molecule strategies to increase brain sRAGE levels, offering a strategic alternative to the challenges posed by the complexity of AD.
Collapse
Affiliation(s)
- Seung‐Hyun Baek
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Suji Hong
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Eunae Kim
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Sunyoung Park
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Minyoung Lee
- Department of Molecular Science and TechnologyAjou UniversitySuwon16499Republic of Korea
| | - Jinsu Park
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Yoonsuk Cho
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | | | | | - Youngkwang Yun
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
| | - Youbin Kim
- School of Biological SciencesSeoul National UniversitySeoul08826Republic of Korea
| | | | - Keunsoo Kang
- Deargen Inc.Daejeon34051Republic of Korea
- Department of MicrobiologyCollege of Science and TechnologyDankook UniversityCheonan31116Republic of Korea
| | - Sangyong Jung
- Department of Medical ScienceCollege of MedicineCHA UniversitySeongnam13496Republic of Korea
| | - Jun Pyo Kim
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoul06355Republic of Korea
| | - Eunha Kim
- Department of Molecular Science and TechnologyAjou UniversitySuwon16499Republic of Korea
| | - Sang Won Seo
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoul06355Republic of Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterSeoul06351Republic of Korea
- Neuroscience CenterSamsung Medical CenterSeoul06351Republic of Korea
| | - Yong‐Keun Jung
- School of Biological SciencesSeoul National UniversitySeoul08826Republic of Korea
| | - Dong‐Gyu Jo
- School of PharmacySungkyunkwan UniversitySuwon16419Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS)Suwon16419Republic of Korea
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06355Republic of Korea
- Institute of Quantum BiophysicsSungkyunkwan UniversitySuwon16419Republic of Korea
| |
Collapse
|
9
|
Howard AJ, Rim EY, Garrett OD, Shim Y, Notwell JH, Ronald PC. Combining Directed Evolution with Machine Learning Enables Accurate Genotype-to-Phenotype Predictions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635131. [PMID: 39974914 PMCID: PMC11838293 DOI: 10.1101/2025.01.27.635131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Linking sequence variation to phenotypic effects is critical for efficient exploitation of large genomic datasets. Here we present a novel approach combining directed evolution with protein language modeling to characterize naturally-evolved variants of a rice immune receptor. Using high-throughput directed evolution, we engineered the rice immune receptor Pik-1 to bind and recognize the fungal proteins Avr-PikC and Avr-PikF, which evade detection by currently characterized Pik-1 alleles. A protein language model was fine-tuned on this data to correlate sequence variation with ligand binding behavior. This modeling was then used to characterize Pik-1 variants found in the 3,000 Rice Genomes Project dataset. Two variants scored highly for binding against Avr-PikC, and in vitro analyses confirmed their improved ligand binding over the wild-type Pik-1 receptor. Overall, this machine learning approach identified promising sources of disease resistance in rice and shows potential utility for exploring the phenotypic variation of other proteins of interest.
Collapse
Affiliation(s)
- Alexander J. Howard
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
| | - Ellen Y. Rim
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
| | - Oscar D. Garrett
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
| | - Yejin Shim
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
| | - James H. Notwell
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
| | - Pamela C. Ronald
- Department of Plant Pathology and the Genome Center, University of California, Davis, CA, 95616, USA
- Joint BioEnergy Institute, Emeryville, CA 94608, USA
- Innovative Genomics Institute (IGI), University of California, Berkeley, CA 94720, USA
| |
Collapse
|
10
|
Xu G, Song Y, Jin H, Shi P, Jiao Y, Cao F, Pang J, Sun Y, Fang L, Xia X, Zhao J. Molecular Engineering of a Tumor-Targeting Thione-Derived Diketopyrrolopyrrole Photosensitizer to Attain NIR Excitation Over 850 nm for Efficient Dual Phototherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407727. [PMID: 39413367 PMCID: PMC11615798 DOI: 10.1002/advs.202407727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/22/2024] [Indexed: 10/18/2024]
Abstract
Photosensitizers with near-infrared (NIR) excitation, especially above 800 nm which is highly desired for phototherapy, remain rare due to the fast nonradiative relaxation process induced by exciton-vibration coupling. Here, a diketopyrrolopyrrole-derived photosensitizer (DTPA-S) is developed via thionation of carbonyl groups within the diketopyrrolopyrrole skeleton, which results in a large bathochromic shift of 81 nm, endowing the photosensitizer with strong NIR absorption at 712 nm. DTPA-S is then introduced with a functional biomolecule (N3-PEG2000-RGD) via click reaction for the construction of integrin αvβ3 receptor-targeted nano-micelles (NanoDTPA-S/RGD), which endows the photosensitizer with a further superlarge absorption redshift of 138 nm, thus extending the absorption maxima to ≈850 nm. Remarkably, thiocarbonyl substitution increases the nonbonding characters in frontier molecular orbitals, which can effectively suppress the nonradiative vibrational relaxation process via reducing the reorganization energy, enabling efficient reactive oxygen species (ROS) generation under 880 nm excitation. Screened by in vitro and in vivo assays, NanoDTPA-S/RGD with high water solubility, excellent tumor-targeting ability, and photodynamic/photothermal therapy synergistic effect exhibits satisfactory phototherapeutic performance. Overall, this study demonstrates a new design of efficient NIR-triggered diketopyrrolopyrrole photosensitizer with facile installation of functional biomolecules for potential clinical applications.
Collapse
Affiliation(s)
- Gang Xu
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Yunxia Song
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Haifeng Jin
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Pengmin Shi
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Yubo Jiao
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Fangzhou Cao
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Jie Pang
- State Key Lab of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yanyan Sun
- School of Chemistry and Life SciencesSuzhou University of Science and TechnologySuzhou215009China
| | - Lei Fang
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| | - Xing‐Hua Xia
- State Key Lab of Analytical Chemistry for Life ScienceSchool of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Jian Zhao
- Jiangsu Province Hi‐Tech Key Laboratory for Biomedical ResearchSchool of Chemistry and Chemical EngineeringSoutheast UniversityNanjing211189China
| |
Collapse
|
11
|
Tan Y, Zhao Z, Han Q, Xu P, Shen X, Jiang Y, Xu Q, Wu X. Identification of an RNA-binding perturbing characteristic for thiopurine drugs and their derivatives to disrupt CELF1-RNA interaction. Nucleic Acids Res 2024; 52:10810-10822. [PMID: 39268573 PMCID: PMC11472155 DOI: 10.1093/nar/gkae788] [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: 03/26/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
RNA-binding proteins (RBPs) are attractive targets in human pathologies. Despite a number of efforts to target RBPs with small molecules, it is still difficult to develop RBP inhibitors, asking for a deeper understanding of how to chemically perturb RNA-binding activity. In this study, we found that the thiopurine drugs (6-mercaptopurine and 6-thioguanine) effectively disrupt CELF1-RNA interaction. The disrupting activity relies on the formation of disulfide bonds between the thiopurine drugs and CELF1. Mutating the cysteine residue proximal to the RNA recognition motifs (RRMs), or adding reducing agents, abolishes the disrupting activity. Furthermore, the 1,2,4-triazole-3-thione, a thiopurine analogue, was identified with 20-fold higher disrupting activity. Based on this analogue, we found that compound 9 disrupts CELF1-RNA interaction in living cells and ameliorates CELF1-mediated myogenesis deficiency. In summary, we identified a thiol-mediated binding mechanism for thiopurine drugs and their derivatives to perturb protein-RNA interaction, which provides novel insight for developing RBP inhibitors. Additionally, this work may benefit the pharmacological and toxicity research of thiopurine drugs.
Collapse
Affiliation(s)
- Yang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhibo Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Qingfang Han
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Peipei Xu
- Department of Hematology, Drum Tower Hospital Affiliated to Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaopeng Shen
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yajun Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xingxin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
12
|
Meena JK, Wang JH, Neill NJ, Keough D, Putluri N, Katsonis P, Koire AM, Lee H, Bowling EA, Tyagi S, Orellana M, Dominguez-Vidaña R, Li H, Eagle K, Danan C, Chung HC, Yang AD, Wu W, Kurley SJ, Ho BM, Zoeller JR, Olson CM, Meerbrey KL, Lichtarge O, Sreekumar A, Dacso CC, Guddat LW, Rejman D, Hocková D, Janeba Z, Simon LM, Lin CY, Pillon MC, Westbrook TF. MYC Induces Oncogenic Stress through RNA Decay and Ribonucleotide Catabolism in Breast Cancer. Cancer Discov 2024; 14:1699-1716. [PMID: 39193992 PMCID: PMC11372365 DOI: 10.1158/2159-8290.cd-22-0649] [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: 07/21/2022] [Revised: 09/12/2023] [Accepted: 05/06/2024] [Indexed: 08/29/2024]
Abstract
Upregulation of MYC is a hallmark of cancer, wherein MYC drives oncogenic gene expression and elevates total RNA synthesis across cancer cell transcriptomes. Although this transcriptional anabolism fuels cancer growth and survival, the consequences and metabolic stresses induced by excess cellular RNA are poorly understood. Herein, we discover that RNA degradation and downstream ribonucleotide catabolism is a novel mechanism of MYC-induced cancer cell death. Combining genetics and metabolomics, we find that MYC increases RNA decay through the cytoplasmic exosome, resulting in the accumulation of cytotoxic RNA catabolites and reactive oxygen species. Notably, tumor-derived exosome mutations abrogate MYC-induced cell death, suggesting excess RNA decay may be toxic to human cancers. In agreement, purine salvage acts as a compensatory pathway that mitigates MYC-induced ribonucleotide catabolism, and inhibitors of purine salvage impair MYC+ tumor progression. Together, these data suggest that MYC-induced RNA decay is an oncogenic stress that can be exploited therapeutically. Significance: MYC is the most common oncogenic driver of poor-prognosis cancers but has been recalcitrant to therapeutic inhibition. We discovered a new vulnerability in MYC+ cancer where MYC induces cell death through excess RNA decay. Therapeutics that exacerbate downstream ribonucleotide catabolism provide a therapeutically tractable approach to TNBC (Triple-negative Breast Cancer) and other MYC-driven cancers.
Collapse
Affiliation(s)
- Jitendra K. Meena
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Jarey H. Wang
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Nicholas J. Neill
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Dianne Keough
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Amanda M. Koire
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Hyemin Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Elizabeth A. Bowling
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Siddhartha Tyagi
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Mayra Orellana
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Rocio Dominguez-Vidaña
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Heyuan Li
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Kenneth Eagle
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Charles Danan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Hsiang-Ching Chung
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Andrew D. Yang
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - William Wu
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Sarah J. Kurley
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Brian M. Ho
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Joseph R. Zoeller
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas.
| | - Calla M. Olson
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Kristen L. Meerbrey
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Clifford C. Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Luke W. Guddat
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Dana Hocková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Zlatko Janeba
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Lukas M. Simon
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
| | - Charles Y. Lin
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| | - Monica C. Pillon
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
| | - Thomas F. Westbrook
- Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, Texas.
- Verna & Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
13
|
Bayoumy AB, Ansari AR, Mulder CJJ, Schmiegelow K, Florin T, De Boer NKH. Innovating Thiopurine Therapeutic Drug Monitoring: A Systematic Review and Meta-Analysis on DNA-Thioguanine Nucleotides (DNA-TG) as an Inclusive Biomarker in Thiopurine Therapy. Clin Pharmacokinet 2024; 63:1089-1109. [PMID: 39031224 PMCID: PMC11343975 DOI: 10.1007/s40262-024-01393-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND AND OBJECTIVE Thioguanine (TG), azathioprine (AZA), and mercaptopurine (MP) are thiopurine prodrugs commonly used to treat diseases, such as leukemia and inflammatory bowel disease (IBD). 6-thioguanine nucleotides (6-TGNs) have been commonly used for monitoring treatment. High levels of 6-TGNs in red blood cells (RBCs) have been associated with leukopenia, the cutoff levels that predict this side effect remain uncertain. Thiopurines are metabolized and incorporated into leukocyte DNA. Measuring levels of DNA-incorporated thioguanine (DNA-TG) may be a more suitable method for predicting clinical response and toxicities such as leukopenia. Unfortunately, most methodologies to assay 6-TGNs are unable to identify the impact of NUDT15 variants, effecting mostly ethnic populations (e.g., Chinese, Indian, Malay, Japanese, and Hispanics). DNA-TG tackles this problem by directly measuring thioguanine in the DNA, which can be influenced by both TPMT and NUDT15 variants. While RBC 6-TGN concentrations have traditionally been used to optimize thiopurine therapy due to their ease and affordability of measurement, recent developments in liquid chromatography-tandem mass spectrometry (LC-MS/MS) techniques have made measuring DNA-TG concentrations in lymphocytes accurate, reproducible, and affordable. The objective of this systematic review was to assess the current evidence of DNA-TG levels as marker for thiopurine therapy, especially with regards to NUDT15 variants. METHODS A systematic review and meta-analysis were performed on the current evidence for DNA-TG as a marker for monitoring thiopurine therapy, including methods for measurement and the illustrative relationship between DNA-TG and various gene variants (such as TPMT, NUDT15, ITPA, NT5C2, and MRP4). PubMed and Embase were systematically searched up to April 2024 for published studies, using the keyword "DNA-TG" with MeSH terms and synonyms. The electronic search strategy was augmented by a manual examination of references cited in articles, recent reviews, editorials, and meta-analyses. A meta-analysis was performed using R studio 4.1.3. to investigate the difference between the coefficients (Fisher's z-transformed correlation coefficient) of DNA-TG and 6-TGNs levels. A meta-analysis was performed using RevMan version 5.4 to investigate the difference in DNA-TG levels between patients with or without leukopenia using randomized effect size model. The risk of bias was assessed using the Newcastle-Ottowa quality assessment scale. RESULTS In this systematic review, 21 studies were included that measured DNA-TG levels in white blood cells for either patients with ALL (n = 16) or IBD (n = 5). In our meta-analysis, the overall mean difference between patients with leukopenia (ALL + IBD) versus no leukopenia was 134.15 fmol TG/µg DNA [95% confidence interval (CI) (83.78-184.35), P < 0.00001; heterogeneity chi squared of 5.62, I2 of 47%]. There was a significant difference in DNA-TG levels for patients with IBD with and without leukopenia [161.76 fmol TG/µg DNA; 95% CI (126.23-197.29), P < 0.00001; heterogeneity chi squared of 0.20, I2 of 0%]. No significant difference was found in DNA-TG level between patients with ALL with or without leukopenia (57.71 fmol TG/µg DNA [95% CI (- 22.93 to 138.35), P < 0.80]). DNA-TG monitoring was found to be a promising method for predicting relapse rates in patients with ALL, and DNA-TG levels are likely a better predictor for leukopenia in patients with IBD than RBC 6-TGNs levels. DNA-TG levels have been shown to correlate with various gene variants (TPMT, NUDT15, ITPA, and MRP4) in various studies, points to its potential as a more informative marker for guiding thiopurine therapy across diverse genetic backgrounds. CONCLUSIONS This systematic review strongly supports the further investigation of DNA-TG as a marker for monitoring thiopurine therapy. Its correlation with treatment outcomes, such as relapse-free survival in ALL and the risk of leukopenia in IBD, underscores its role in enhancing personalized treatment approaches. DNA-TG effectively identifies NUDT15 variants and predicts late leukopenia in patients with IBD, regardless of their NUDT15 variant status. The recommended threshold for late leukopenia prediction in patients with IBD with DNA-TG is suggested to be between 320 and 340 fmol/µg DNA. More clinical research on DNA-TG implementation is mandatory to improve patient care and to improve inclusivity in thiopurine treatment.
Collapse
Affiliation(s)
- Ahmed B Bayoumy
- Department of Internal Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, The Netherlands.
| | - A R Ansari
- Department of Gastroenterology and Hepatology, London Bridge Hospital, London, UK
| | - C J J Mulder
- Department of Gastroenterology and Hepatology, AGEM Research Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - K Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The Juliane Marie Centre, The University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, The Faculty of Health Sciences, The University of Copenhagen, Copenhagen, Denmark
| | - Timothy Florin
- Mater Research, University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - N K H De Boer
- Department of Gastroenterology and Hepatology, AGEM Research Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Maillard M, Nishii R, Yang W, Hoshitsuki K, Chepyala D, Lee SHR, Nguyen JQ, Relling MV, Crews KR, Leggas M, Singh M, Suang JLY, Yeoh AEJ, Jeha S, Inaba H, Pui CH, Karol SE, Trehan A, Bhatia P, Antillon Klussmann FG, Bhojwani D, Haidar CE, Yang JJ. Additive effects of TPMT and NUDT15 on thiopurine toxicity in children with acute lymphoblastic leukemia across multiethnic populations. J Natl Cancer Inst 2024; 116:702-710. [PMID: 38230823 PMCID: PMC11077315 DOI: 10.1093/jnci/djae004] [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: 11/05/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Thiopurines such as mercaptopurine (MP) are widely used to treat acute lymphoblastic leukemia (ALL). Thiopurine-S-methyltransferase (TPMT) and Nudix hydrolase 15 (NUDT15) inactivate thiopurines, and no-function variants are associated with drug-induced myelosuppression. Dose adjustment of MP is strongly recommended in patients with intermediate or complete loss of activity of TPMT and NUDT15. However, the extent of dosage reduction recommended for patients with intermediate activity in both enzymes is currently not clear. METHODS MP dosages during maintenance were collected from 1768 patients with ALL in Singapore, Guatemala, India, and North America. Patients were genotyped for TPMT and NUDT15, and actionable variants defined by the Clinical Pharmacogenetics Implementation Consortium were used to classify patients as TPMT and NUDT15 normal metabolizers (TPMT/NUDT15 NM), TPMT or NUDT15 intermediate metabolizers (TPMT IM or NUDT15 IM), or TPMT and NUDT15 compound intermediate metabolizers (TPMT/NUDT15 IM/IM). In parallel, we evaluated MP toxicity, metabolism, and dose adjustment using a Tpmt/Nudt15 combined heterozygous mouse model (Tpmt+/-/Nudt15+/-). RESULTS Twenty-two patients (1.2%) were TPMT/NUDT15 IM/IM in the cohort, with the majority self-reported as Hispanics (68.2%, 15/22). TPMT/NUDT15 IM/IM patients tolerated a median daily MP dose of 25.7 mg/m2 (interquartile range = 19.0-31.1 mg/m2), significantly lower than TPMT IM and NUDT15 IM dosage (P < .001). Similarly, Tpmt+/-/Nudt15+/- mice displayed excessive hematopoietic toxicity and accumulated more metabolite (DNA-TG) than wild-type or single heterozygous mice, which was effectively mitigated by a genotype-guided dose titration of MP. CONCLUSION We recommend more substantial dose reductions to individualize MP therapy and mitigate toxicity in TPMT/NUDT15 IM/IM patients.
Collapse
Affiliation(s)
- Maud Maillard
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rina Nishii
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Keito Hoshitsuki
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Divyabharathi Chepyala
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Shawn H R Lee
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jenny Q Nguyen
- Personalized Care Program, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Mary V Relling
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Kristine R Crews
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mark Leggas
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Meenu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Joshua L Y Suang
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Allen E J Yeoh
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sima Jeha
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hiroto Inaba
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Amita Trehan
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cyrine E Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
15
|
Pal AK, Datta A. First-principles design of heavy-atom-free singlet oxygen photosensitizers for photodynamic therapy. J Chem Phys 2024; 160:164720. [PMID: 38682739 DOI: 10.1063/5.0196557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/10/2024] [Indexed: 05/01/2024] Open
Abstract
In photodynamic therapy (PDT) treatment, heavy-atom-free photosensitizers (PSs) are a great source of singlet oxygen photosensitizer. Reactive oxygen species (ROS) are produced by an energy transfer from the lowest energy triplet excited state to the molecular oxygen of cancer cells. To clarify the photophysical characteristics in the excited states of a few experimentally identified thionated (>C=S) molecules and their oxygenated congeners (>C=O), a quantum chemical study is conducted. This study illustrates the properties of the excited states in oxygen congeners that render them unsuitable for PDT treatment. Concurrently, a hierarchy is presented based on the utility of the lowest-energy triplet excitons of thionated compounds. Their non-radiative decay rates are calculated for reverse-ISC and inter-system crossover (ISC) processes. In addition, the vibronic importance of C=O and C=S bonds is clarified by the computation of the Huang-Rhys factor, effective vibrational mode, and reorganization energy inside the Marcus-Levich-Jörtner system. ROS generation in thionated PSs exceeds their oxygen congeners as kf ≪ kISC, where radiative decay rate is designated as kf. As a result, the current work offers a calculated strategy for analyzing the effectiveness of thionated photosensitizers in PDT.
Collapse
Affiliation(s)
- Arun K Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Ayan Datta
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| |
Collapse
|
16
|
Massey JC, Magagnoli J, Sutton SS, Buckhaults PJ, Wyatt MD. Collateral damage of NUDT15 deficiency in cancer provides a cancer pharmacogenetic therapeutic window with thiopurines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588560. [PMID: 38645136 PMCID: PMC11030356 DOI: 10.1101/2024.04.08.588560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Genome instability is a hallmark of cancer and are driven by mutations in oncogenes and tumor suppressor genes. Despite successes seen with select targeted therapeutics, this type of personalized medicine is only beneficial for a small subpopulation of cancer patients who have one of a few actionable genetic changes. Most tumors also contain hundreds of passenger mutations that offered no fitness advantage or disadvantage during tumor evolution. Mutations in known pharmacogenetic (PGx) loci for which germline variants encode variability in drug response can cause somatically acquired drug sensitivity. The NUDT15 gene is a known PGx locus that participates in the rate-limiting metabolism of thiopurines. People with two defective germline alleles of NUDT15 are hypersensitive to the toxic effects of thiopurines. NUDT15 is located adjacent to the Retinoblastoma ( RB1 ) tumor suppressor gene, which often undergoes homozygous deletion in retinoblastomas and other epithelial cancers. We observed that RB1 undergoes homozygous deletions in 9.4% of prostate adenocarcinomas and 2.5% of ovarian cancers, and in nearly all of these cases NUDT15 is also lost. Moreover, 44% of prostate adenocarcinomas and over 60% of ovarian cancers have lost one allele of NUDT15, which predicts that a majority of all prostate and ovarian cancers have somatically acquired hypersensitivity to thiopurine treatment. We performed a retrospective analysis of >16,000 patients in the US Veterans Administration health care system and found concurrent xanthine oxidase inhibition (XOi) and thiopurine usage for non-cancer indications is significantly associated with reduced incidence of prostate cancer. The hazard ratio for the development of prostate cancer in patients treated with thiopurines and XOi was 0.562 (0.301-1.051) for the unmatched cohort and 0.389 (0.185-0.819) for the propensity score matched cohort. We experimentally depleted NUDT15 from ovarian and prostate cancer cell lines and observed a dramatic sensitization to thiopurine-induced and DNA damage-dependent toxicity. These results indicate that somatic loss of NUDT15 predicts therapeutic sensitivity to a low cost and well tolerated drug with a broad therapeutic window.
Collapse
|
17
|
Murt A, Bayram B, Yılmaz U, Seyahi N, Eşkazan AE. Chronic Myeloid Leukemia in Renal Transplantation Patients in the Era of Tyrosine Kinase Inhibitors: A Case Report and Review of the Literature. Nephron Clin Pract 2024; 148:563-568. [PMID: 38574488 DOI: 10.1159/000538532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024] Open
Abstract
Lifelong immunosuppression, cytotoxic effects of some immunosuppressive drugs, and opportunistic oncogenic viruses increase malignancy risks in solid organ recipients. The risk of myeloid neoplasms including chronic myeloid leukemia (CML) is also increased in this patient population. Tyrosine kinase inhibitors (TKIs), the key element of CML therapy, should be used cautiously in transplantation patients as they may interact with calcineurin inhibitors. With this report, a 63-year-old female kidney transplant recipient who developed CML 9 years after kidney transplantation is presented. CML in this patient was treated with a slightly reduced dose of imatinib (300 mg) due to concerns of adverse events including its interaction with tacrolimus. Deep molecular response (DMR) was achieved at 12 months under imatinib treatment. The patient is still in DMR after 30 months of follow-up, and she did not experience any adverse events or acute rejection episodes. CML and the use of TKIs in kidney transplant patients have been discussed with an extensive literature review. In this patient population, TKIs are generally well tolerated with achievement of treatment responses and good prognosis. Graft functions are also well maintained as long as drug interactions are monitored.
Collapse
Affiliation(s)
- Ahmet Murt
- Division of Nephrology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Batuhan Bayram
- Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Umut Yılmaz
- Division of Hematology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Nurhan Seyahi
- Division of Nephrology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
18
|
Bentley BH, Cecil AE, Lippert WC. Atypical Intrahepatic Cholestasis of Pregnancy: A Red Herring. ACG Case Rep J 2024; 11:e01339. [PMID: 38638198 PMCID: PMC11025705 DOI: 10.14309/crj.0000000000001339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
Intrahepatic cholestasis of pregnancy is an intrahepatic etiology of acute cholestasis commonly defined by pruritus and increased bile acids, liver transaminases, and, occasionally, bilirubin. Azathioprine is an immunosuppressive agent associated with various forms of hepatoxicity, ranging from transient rises in serum aminotransferase levels, acute cholestatic injury, and chronic hepatic injury. In this report, we present a 20-year-old pregnant woman who presented with cholestatic liver injury due to intrahepatic cholestasis of pregnancy with a clinical picture complicated by increased levels of azathioprine metabolites.
Collapse
Affiliation(s)
- Blake H. Bentley
- Department of Internal Medicine. Wake Forest University School of Medicine, Winston-Salem, NC
| | - Alexa E. Cecil
- Department of Internal Medicine. Wake Forest University School of Medicine, Winston-Salem, NC
| | - William C. Lippert
- Department of Internal Medicine. Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
19
|
Sato R, Yamada Y, Kashihara W, Nishihara T, Tanabe K, Xu YZ, Suzuki T. Excited state properties of 5-fluoro-4-thiouridine derivative †. Photochem Photobiol 2024; 100:434-442. [PMID: 38312100 DOI: 10.1111/php.13918] [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: 09/26/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
The excited state properties of thionated 5-fluorouridine (2',3',5'-tri-O-acetyl-5-fluoro-4-thiouridine; ta5F4TUrd), synthesized with Lawesson's reagent, have been intensively investigated with nanosecond transient absorption spectroscopy, time-resolved thermal lensing, near-infrared emission, and quantum chemical calculation. The intrinsic triplet lifetime of ta5F4TUrd was determined to be4.2 ± 0.7 μs in acetonitrile, and the formation quantum yield of the excited triplet state was as large as0.79 ± 0.01 . The quenching rate constants of the triplet ta5F4TUrd by the dissolved oxygen molecule and by the self-quenching process were found to be nearly equal to the diffusion-controlled rate of acetonitrile. The quantum yield of the singlet molecular oxygen produced through energy transfer between the triplet ta5F4TUrd and the dissolved oxygen,Φ Δ , was successfully determined to be0.61 ± 0.02 under the oxygen-saturated condition. From the oxygen concentration dependence of theΦ Δ value, the fraction of triplet ta5F4TUrd quenched by dissolved oxygen which gives rise to the 1 O2 * formation,S Δ , was successfully obtained to be0.78 ± 0.01 , which was the largest among the thionucleobases and the thionucleosides reported so far. This could be due to the lower energy and/or the ππ* character of the triplet state.
Collapse
Affiliation(s)
- Rin Sato
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| | - Yoshino Yamada
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| | - Wataru Kashihara
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| | - Tatsuya Nishihara
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| | - Kazuhito Tanabe
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| | - Yao-Zhong Xu
- School of Life, Health and Chemical Sciences, The Open University, Keynes, UK
| | - Tadashi Suzuki
- Department of Chemistry and Biological Science, Aoyama Gakuin University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
20
|
Bayoumy AB, Mulder CJJ, Ansari AR, Barclay ML, Florin T, Kiszka-Kanowitz M, Derijks L, Sharma V, de Boer NKH. Uphill battle: Innovation of thiopurine therapy in global inflammatory bowel disease care. Indian J Gastroenterol 2024; 43:36-47. [PMID: 38383877 PMCID: PMC10924016 DOI: 10.1007/s12664-024-01529-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract that encompasses two major conditions: Crohn's disease (CD) and ulcerative colitis (UC). Historically, IBD has been primarily reported in western countries, but over the past decades, its prevalence is rapidly increasing, especially in lower and middle-income countries (LMICs) such as India and China and also in Sub-Saharan Africa. The prevalence of IBD in LMICs has been the subject of growing concern due to the impact of access to public healthcare and the burden it places on healthcare resources. The classical thiopurines face significant challenges due to cessation of therapy in approximately half of patients within one year due to side effects or ineffectiveness. In this article, we highlight innovating thiopurine treatment for IBD patients in downregulating side effects and improving efficacy.
Collapse
Affiliation(s)
- Ahmed B Bayoumy
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Chris J J Mulder
- Department of Gastroenterology and Hepatology, AGEM Research Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Azhar R Ansari
- Department of Gastroenterology and Hepatology, London Bridge Hospital, London, UK
| | - Murray L Barclay
- Department of Gastroenterology, Christchurch Hospital, Christchurch, Waitaha - Canterbury, New Zealand
- Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, Waitaha - Canterbury, New Zealand
| | - Tim Florin
- Mater Research, University of Queensland, Translational Research Institute, South Brisbane, Australia
| | - Marianne Kiszka-Kanowitz
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre, Denmark
| | - Luc Derijks
- Department of Clinical Pharmacy, Máxima Medical Center, Veldhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Vishal Sharma
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, India
| | - Nanne K H de Boer
- Department of Gastroenterology and Hepatology, AGEM Research Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Rios-Usuga C, Martinez-Gutierrez M, Ruiz-Saenz J. Antiviral Potential of Azathioprine and Its Derivative 6- Mercaptopurine: A Narrative Literature Review. Pharmaceuticals (Basel) 2024; 17:174. [PMID: 38399389 PMCID: PMC10892228 DOI: 10.3390/ph17020174] [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: 12/14/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The use of azathioprine (AZA) in human medicine dates back to research conducted in 1975 that led to the development of several drugs, including 6-mercaptopurine. In 1958, it was shown that 6-mercaptopurine decreased the production of antibodies against earlier administered antigens, raising the hypothesis of an immunomodulatory effect. AZA is a prodrug that belongs to the thiopurine group of drugs that behave as purine analogs. After absorption, it is converted into 6-mercaptopurine. Subsequently, it can be degraded through various enzymatic pathways into inactive compounds and biologically active compounds related to the mechanism of action, which has been the subject of study to evaluate a possible antiviral effect. This study aims to examine the metabolism, mechanism of action, and antiviral potential of AZA and its derivatives, exploring AZA impact on antiviral targets and adverse effects through a narrative literature review. Ultimately, the review will provide insights into the antiviral mechanism, present evidence of its in vitro effectiveness against various DNA and RNA viruses, and suggest in vivo studies to further demonstrate its antiviral effects.
Collapse
Affiliation(s)
- Carolina Rios-Usuga
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (C.R.-U.); (M.M.-G.)
| | - Marlen Martinez-Gutierrez
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (C.R.-U.); (M.M.-G.)
- Grupo de Investigación en Microbiología Veterinaria, Escuela de Microbiología, Universidad de Antioquia UdeA, Medellín 050001, Colombia
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680002, Colombia; (C.R.-U.); (M.M.-G.)
| |
Collapse
|
22
|
Yankilevich P, Nazerai L, Willis SC, Schmiegelow K, De Zio D, Nielsen M. An analysis pipeline for understanding 6-thioguanine effects on a mouse tumour genome. Cancer Immunol Immunother 2024; 73:22. [PMID: 38279992 PMCID: PMC10821971 DOI: 10.1007/s00262-023-03610-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/07/2023] [Indexed: 01/29/2024]
Abstract
Mouse tumour models are extensively used as a pre-clinical research tool in the field of oncology, playing an important role in anticancer drugs discovery. Accordingly, in cancer genomics research, the demand for next-generation sequencing (NGS) is increasing, and consequently, the need for data analysis pipelines is likewise growing. Most NGS data analysis solutions to date do not support mouse data or require highly specific configuration for their use. Here, we present a genome analysis pipeline for mouse tumour NGS data including the whole-genome sequence (WGS) data analysis flow for somatic variant discovery, and the RNA-seq data flow for differential expression, functional analysis and neoantigen prediction. The pipeline is based on standards and best practices and integrates mouse genome references and annotations. In a recent study, the pipeline was applied to demonstrate the efficacy of low dose 6-thioguanine (6TG) treatment on low-mutation melanoma in a pre-clinical mouse model. Here, we further this study and describe in detail the pipeline and the results obtained in terms of tumour mutational burden (TMB) and number of predicted neoantigens, and correlate these with 6TG effects on tumour volume. Our pipeline was expanded to include a neoantigen analysis, resulting in neopeptide prediction and MHC class I antigen presentation evaluation. We observed that the number of predicted neoepitopes were more accurate indicators of tumour immune control than TMB. In conclusion, this study demonstrates the usability of the proposed pipeline, and suggests it could be an essential robust genome analysis platform for future mouse genomic analysis.
Collapse
Affiliation(s)
- Patricio Yankilevich
- Bioinformatics Core Facility, Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - Loulieta Nazerai
- Melanoma Research Team, Danish Cancer Institute, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Shona Caroline Willis
- Melanoma Research Team, Danish Cancer Institute, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Danish Cancer Institute, Copenhagen, Denmark
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morten Nielsen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
23
|
Robello M, Nikolayevskiy H, Scerba MT, Nahui Palomino RA, Mercurio V, Appella DH. Prodrug Strategy Extends the Use of Anti-HIV Sulfanylbenzamides for Application In Vivo. ACS Pharmacol Transl Sci 2024; 7:259-273. [PMID: 38250006 PMCID: PMC10795369 DOI: 10.1021/acsptsci.3c00260] [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: 09/29/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Abstract
Sulfanylbenzamide thioesters are molecules with anti-HIV activity that disrupt zinc coordination in the viral protein NCp7. These molecules are useful as topical microbicides; however, they are too unstable to be used systemically. In this article, a nitroimidazole prodrug was used to protect the sulfanylbenzamide to convey blood stability and oral bioavailability to the molecule. Studies on the molecule called nipamovir were performed to assess the rate of prodrug cleavage, antiviral activity, mechanism of metabolism, and in vivo pharmacokinetics in several different species. An efficient and inexpensive synthesis of nipamovir is also described. The results indicate that nipamovir could be further developed as a new type of drug to treat HIV infection.
Collapse
Affiliation(s)
- Marco Robello
- Synthetic Bioactive Molecules Section, Laboratory of
Bioorganic Chemistry (LBC), National Institute of Diabetes and Digestive and Kidney
Diseases (NIDDK), National Institutes of Health, 8 Center
Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Herman Nikolayevskiy
- Synthetic Bioactive Molecules Section, Laboratory of
Bioorganic Chemistry (LBC), National Institute of Diabetes and Digestive and Kidney
Diseases (NIDDK), National Institutes of Health, 8 Center
Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Michael T. Scerba
- Synthetic Bioactive Molecules Section, Laboratory of
Bioorganic Chemistry (LBC), National Institute of Diabetes and Digestive and Kidney
Diseases (NIDDK), National Institutes of Health, 8 Center
Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Rogers Alberto Nahui Palomino
- Section on Intercellular Interactions, Eunice Kennedy
Shriver National Institute of Child Health and Human Development, National
Institutes of Health, Bethesda, Maryland 20892, United
States
| | - Vincenzo Mercurio
- Section on Intercellular Interactions, Eunice Kennedy
Shriver National Institute of Child Health and Human Development, National
Institutes of Health, Bethesda, Maryland 20892, United
States
| | - Daniel H. Appella
- Synthetic Bioactive Molecules Section, Laboratory of
Bioorganic Chemistry (LBC), National Institute of Diabetes and Digestive and Kidney
Diseases (NIDDK), National Institutes of Health, 8 Center
Drive, Room 404, Bethesda, Maryland 20892, United States
| |
Collapse
|
24
|
Rana N, Grover P, Singh H. Recent Developments and Future Perspectives of Purine Derivatives as a Promising Scaffold in Drug Discovery. Curr Top Med Chem 2024; 24:541-579. [PMID: 38288806 DOI: 10.2174/0115680266290152240110074034] [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: 10/30/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 05/31/2024]
Abstract
Numerous purine-containing compounds have undergone extensive investigation for their medical efficacy across various diseases. The swift progress in purine-based medicinal chemistry has brought to light the therapeutic capabilities of purine-derived compounds in addressing challenging medical conditions. Defined by a heterocyclic ring comprising a pyrimidine ring linked with an imidazole ring, purine exhibits a diverse array of therapeutic attributes. This review systematically addresses the multifaceted potential of purine derivatives in combating various diseases, including their roles as anticancer agents, antiviral compounds (anti-herpes, anti-HIV, and anti-influenzae), autoimmune and anti-inflammatory agents, antihyperuricemic and anti-gout solutions, antimicrobial agents, antitubercular compounds, anti-leishmanial agents, and anticonvulsants. Emphasis is placed on the remarkable progress made in developing purine-based compounds, elucidating their significant target sites. The article provides a comprehensive exploration of developments in both natural and synthetic purines, offering insights into their role in managing a diverse range of illnesses. Additionally, the discussion delves into the structure-activity relationships and biological activities of the most promising purine molecules. The intriguing capabilities revealed by these purine-based scaffolds unequivocally position them at the forefront of drug candidate development. As such, this review holds potential significance for researchers actively involved in synthesizing purine-based drug candidates, providing a roadmap for the continued advancement of this promising field.
Collapse
Affiliation(s)
- Neha Rana
- School of Pharmacy (SOP), Noida International University, Yamuna Expressway, Gautam Budh Nagar, 203201, India
| | - Parul Grover
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - Hridayanand Singh
- Dr. K. N. Modi Institute of Pharmaceutical Education and Research, Modinagar, 201204, Uttar Pradesh, India
| |
Collapse
|
25
|
Salama MM, Aborehab NM, El Mahdy NM, Zayed A, Ezzat SM. Nanotechnology in leukemia: diagnosis, efficient-targeted drug delivery, and clinical trials. Eur J Med Res 2023; 28:566. [PMID: 38053150 DOI: 10.1186/s40001-023-01539-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023] Open
Abstract
Leukemia is a group of malignant disorders which affect the blood and blood-forming tissues in the bone marrow, lymphatic system, and spleen. Many types of leukemia exist; thus, their diagnosis and treatment are somewhat complicated. The use of conventional strategies for treatment such as chemotherapy and radiotherapy may develop many side effects and toxicity. Hence, modern research is concerned with the development of specific nano-formulations for targeted delivery of anti-leukemic drugs avoiding toxic effects on normal cells. Nanostructures can be applied not only in treatment but also in diagnosis. In this article, types of leukemia, its causes, diagnosis as well as conventional treatment of leukemia shall be reviewed. Then, the use of nanoparticles in diagnosis of leukemia and synthesis of nanocarriers for efficient delivery of anti-leukemia drugs being investigated in in vivo and clinical studies. Therefore, it may contribute to the discovery of novel and emerging nanoparticles for targeted treatment of leukemia with less side effects and toxicities.
Collapse
Affiliation(s)
- Maha M Salama
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Suez Desert Road, Cairo, 11837, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Nihal M El Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Ahmed Zayed
- Department of Pharmacognosy, College of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527, Egypt
| | - Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt.
| |
Collapse
|
26
|
Lemberg KM, Ali ES, Krecmerova M, Aguilar JMH, Alt J, Peters DE, Zhao L, Wu Y, Nuha N, Asara JM, Staedtke V, Pratilas CA, Majer P, Rais R, Ben-Sahra I, Slusher BS. Pro-905, a Novel Purine Antimetabolite, Combines with Glutamine Amidotransferase Inhibition to Suppress Growth of Malignant Peripheral Nerve Sheath Tumor. Mol Cancer Ther 2023; 22:1390-1403. [PMID: 37616542 PMCID: PMC10690047 DOI: 10.1158/1535-7163.mct-23-0258] [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: 04/27/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are highly aggressive soft-tissue sarcomas that arise from neural tissues and carry a poor prognosis. Previously, we found that the glutamine amidotransferase inhibitor JHU395 partially impeded tumor growth in preclinical models of MPNST. JHU395 inhibits de novo purine synthesis in human MPNST cells and murine tumors with partial decreases in purine monophosphates. On the basis of prior studies showing enhanced efficacy when glutamine amidotransferase inhibition was combined with the antimetabolite 6-mercaptopurine (6-MP), we hypothesized that such a combination would be efficacious in MPNST. Given the known toxicity associated with 6-MP, we set out to develop a more efficient and well-tolerated drug that targets the purine salvage pathway. Here, we report the discovery of Pro-905, a phosphoramidate protide that delivered the active nucleotide antimetabolite thioguanosine monophosphate (TGMP) to tumors over 2.5 times better than equimolar 6-MP. Pro-905 effectively prevented the incorporation of purine salvage substrates into nucleic acids and inhibited colony formation of human MPNST cells in a dose-dependent manner. In addition, Pro-905 inhibited MPNST growth and was well-tolerated in both human patient-derived xenograft (PDX) and murine flank MPNST models. When combined with JHU395, Pro-905 enhanced the colony formation inhibitory potency of JHU395 in human MPNST cells and augmented the antitumor efficacy of JHU395 in mice. In summary, the dual inhibition of the de novo and purine salvage pathways in preclinical models may safely be used to enhance therapeutic efficacy against MPNST.
Collapse
Affiliation(s)
- Kathryn M. Lemberg
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Johns Hopkins Drug Discovery, Baltimore, Maryland
| | - Eunus S. Ali
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Marcela Krecmerova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Jesse Alt
- Johns Hopkins Drug Discovery, Baltimore, Maryland
| | - Diane E. Peters
- Johns Hopkins Drug Discovery, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Liang Zhao
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ying Wu
- Johns Hopkins Drug Discovery, Baltimore, Maryland
| | - Naziba Nuha
- Johns Hopkins Drug Discovery, Baltimore, Maryland
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard University School of Medicine, Boston, Massachusetts
| | - Verena Staedtke
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Christine A. Pratilas
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Rana Rais
- Johns Hopkins Drug Discovery, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Barbara S. Slusher
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Johns Hopkins Drug Discovery, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Departments of Medicine, Neuroscience, Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
27
|
Gallardo-Cóndor J, Naranjo P, Atarihuana S, Coello D, Guevara-Ramírez P, Flores-Espinoza R, Burgos G, López-Cortés A, Cabrera-Andrade A. Population-Specific Distribution of TPMT Deficiency Variants and Ancestry Proportions in Ecuadorian Ethnic Groups: Towards Personalized Medicine. Ther Clin Risk Manag 2023; 19:1005-1018. [PMID: 38050617 PMCID: PMC10693761 DOI: 10.2147/tcrm.s432856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
Purpose Thiopurine S-methyltransferase (TPMT) is an enzyme that metabolizes purine analogs, agents used in the treatment of acute lymphoblastic leukemia. Improper drug metabolism leads to toxicity in chemotherapy patients and reduces treatment effectiveness. TPMT variants associated with reduced enzymatic activity vary across populations. Therefore, studying these variants in heterogeneous populations, such as Ecuadorians, can help identify molecular causes of deficiency for this enzyme. Methods We sequenced the entire TPMT coding region in 550 Ecuadorian individuals from Afro-Ecuadorian, Indigenous, Mestizo, and Montubio ethnicities. Moreover, we conducted an ancestry analysis using 46 informative ancestry markers. Results We identified 8 single nucleotide variants in the coding region of TPMT. The most prevalent alleles were TPMT*3A, TPMT*3B, and TPMT*3C, with frequencies of 0.055, 0.012, and 0.015, respectively. Additionally, we found rare alleles TPMT*4 and TPMT*8 with frequencies of 0.005 and 0.003. Correlating the ancestry proportions with TPMT-deficient genotypes, we observed that the Native American ancestry proportion influenced the distribution of the TPMT*1/TPMT*3A genotype (OR = 5.977, p = 0.002), while the contribution of African ancestral populations was associated with the TPMT*1/TPMT*3C genotype (OR = 9.769, p = 0.003). The rates of TPMT-deficient genotypes observed in Mestizo (f = 0.121) and Indigenous (f = 0.273) groups provide evidence for the influence of Native American ancestry and the prevalence of the TPMT*3A allele. In contrast, although Afro-Ecuadorian groups demonstrate similar deficiency rates (f = 0.160), the genetic factors involved are associated with contributions from African ancestral populations, specifically the prevalent TPMT*3C allele. Conclusion The distribution of TPMT-deficient variants offers valuable insights into the populations under study, underscoring the necessity for genetic screening strategies to prevent thiopurine toxicity events among Latin American minority groups.
Collapse
Affiliation(s)
| | - Pablo Naranjo
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| | - Sebastián Atarihuana
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| | - Dayana Coello
- Laboratorios de Investigación, Universidad de Las Américas, Quito, Ecuador
| | - Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rodrigo Flores-Espinoza
- Laboratório de Diagnóstico por DNA (LDD), Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Germán Burgos
- One Health Research Group, Facultad de Medicina, Universidad de Las Américas, Quito, Ecuador
- Grupo de Medicina Xenomica, Instituto de Ciencias Forenses, Universidad de Santiago de Compostela, Satiago de Compostela, Spain
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Alejandro Cabrera-Andrade
- Escuela de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
28
|
Ali M, Park IH, Kim J, Kim G, Oh J, You JS, Kim J, Shin JS, Yoon SS. How Deep Learning in Antiviral Molecular Profiling Identified Anti-SARS-CoV-2 Inhibitors. Biomedicines 2023; 11:3134. [PMID: 38137356 PMCID: PMC10740425 DOI: 10.3390/biomedicines11123134] [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: 10/18/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
The integration of artificial intelligence (AI) into drug discovery has markedly advanced the search for effective therapeutics. In our study, we employed a comprehensive computational-experimental approach to identify potential anti-SARS-CoV-2 compounds. We developed a predictive model to assess the activities of compounds based on their structural features. This model screened a library of approximately 700,000 compounds, culminating in the selection of the top 100 candidates for experimental validation. In vitro assays on human intestinal epithelial cells (Caco-2) revealed that 19 of these compounds exhibited inhibitory activity. Notably, eight compounds demonstrated dose-dependent activity in Vero cell lines, with half-maximal effective concentration (EC50) values ranging from 1 μM to 7 μM. Furthermore, we utilized a clustering approach to pinpoint potential nucleoside analog inhibitors, leading to the discovery of two promising candidates: azathioprine and its metabolite, thioinosinic acid. Both compounds showed in vitro activity against SARS-CoV-2, with thioinosinic acid also significantly reducing viral loads in mouse lungs. These findings underscore the utility of AI in accelerating drug discovery processes.
Collapse
Affiliation(s)
- Mohammed Ali
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - In Ho Park
- Department of Biomedical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Junebeom Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Gwanghee Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jooyeon Oh
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jin Sun You
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jieun Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- BioMe Inc., Seoul 02455, Republic of Korea
| |
Collapse
|
29
|
Kim DG, Yim SH, Min EK, Choi MC, Joo DJ, Kim MS, Lee JG. Cumulative exposure to tacrolimus during early period after liver transplantation does not affect the recurrence of hepatocellular carcinoma. Sci Rep 2023; 13:20236. [PMID: 37981643 PMCID: PMC10658176 DOI: 10.1038/s41598-023-46803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/05/2023] [Indexed: 11/21/2023] Open
Abstract
The clinical effects of tacrolimus (TAC) exposure on hepatocellular carcinoma (HCC) recurrence after liver transplantation (LT) remain unclear. In this retrospective single centric study, 512 patients who underwent LT for HCC were divided into four groups according to cumulative exposure to tacrolimus (CET) during 3 months after LT: conventional (n = 218), aggressive minimization (n = 32), minimization (n = 161), and high exposure (n = 101). Impact of CET on HCC recurrence and death were analyzed. Compared with the conventional group, the other three CET groups showed a similar risk of HCC recurrence. The aggressive minimization group showed a higher risk [hazard ratio (HR) 5.64, P < 0.001] and the high exposure group showed a marginal risk (HR 1.67, P = 0.081) of overall death compared to the conventional group. CET during 3 months was not associated with HCC recurrence in the matched cohort and various subgroups. TAC minimization is not effective to prevent HCC recurrence but could result in higher mortality in LT recipients.
Collapse
Affiliation(s)
- Deok-Gie Kim
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Seung Hyuk Yim
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Eun-Ki Min
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Mun Chae Choi
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Dong Jin Joo
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Myoung Soo Kim
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jae Geun Lee
- Department of Surgery, The Research Institute for Transplantation, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
30
|
de Lope-Planelles A, González-Novo R, Madrazo E, Peralta-Carrero G, Cruz Rodríguez MP, Zamora-Carreras H, Torrano V, López-Menéndez H, Roda-Navarro P, Monroy F, Redondo-Muñoz J. Mechanical stress confers nuclear and functional changes in derived leukemia cells from persistent confined migration. Cell Mol Life Sci 2023; 80:316. [PMID: 37801090 PMCID: PMC10558412 DOI: 10.1007/s00018-023-04968-5] [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: 03/14/2023] [Revised: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Nuclear deformability plays a critical role in cell migration. During this process, the remodeling of internal components of the nucleus has a direct impact on DNA damage and cell behavior; however, how persistent migration promotes nuclear changes leading to phenotypical and functional consequences remains poorly understood. Here, we described that the persistent migration through physical barriers was sufficient to promote permanent modifications in migratory-altered cells. We found that derived cells from confined migration showed changes in lamin B1 localization, cell morphology and transcription. Further analysis confirmed that migratory-altered cells showed functional differences in DNA repair, cell response to chemotherapy and cell migration in vivo homing experiments. Experimental modulation of actin polymerization affected the redistribution of lamin B1, and the basal levels of DNA damage in migratory-altered cells. Finally, since major nuclear changes were present in migratory-altered cells, we applied a multidisciplinary biochemical and biophysical approach to identify that confined conditions promoted a different biomechanical response of the nucleus in migratory-altered cells. Our observations suggest that mechanical compression during persistent cell migration has a role in stable nuclear and genomic alterations that might handle the genetic instability and cellular heterogeneity in aging diseases and cancer.
Collapse
Affiliation(s)
- Ana de Lope-Planelles
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Raquel González-Novo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Elena Madrazo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Gracia Peralta-Carrero
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - María Pilar Cruz Rodríguez
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Héctor Zamora-Carreras
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Verónica Torrano
- Department of Biochemistry and Molecular Biology, University of the Basque Country, Leioa, Spain
| | - Horacio López-Menéndez
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, School of Medicine, University Complutense de Madrid and 12 de Octubre Health Research Institute (Imas12) Madrid, Madrid, Spain
| | - Francisco Monroy
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Redondo-Muñoz
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain.
| |
Collapse
|
31
|
Lukaszewicz M, Ferenc-Mrozek A, Kokosza J, Stefaniuk A, Stepinski J, Bojarska E, Darzynkiewicz E. Mammalian Nudt15 hydrolytic and binding activity on methylated guanosine mononucleotides. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:487-495. [PMID: 37644211 PMCID: PMC10618335 DOI: 10.1007/s00249-023-01678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
The Nudt15 enzyme of the NUDIX protein family is the subject of extensive study due to its action on thiopurine drugs used in the treatment of cancer and inflammatory diseases. In addition to thiopurines, Nudt15 is enzymatically active in vitro on several nucleotide substrates. It has also been suggested that this enzyme may play a role in 5'RNA turnover by hydrolyzing m7GDP, a product of mRNA decapping. However, no detailed studies on this substrate with Nudt15 are available. Here, we analyzed the enzymatic activity of Nudt15 with m7GDP, its triphosphate form m7GTP, and the trimethylated counterparts (m32,2,7GDP and m32,2,7GTP). Kinetic data revealed a moderate activity of Nudt15 toward these methylated mononucleotides compared to the dGTP substrate. However m7GDP and m32,2,7GDP showed a distinct stabilization of Nudt15 upon ligand binding, in the same range as dGTP, and thus these two mononucleotides may be used as leading structures in the design of small molecule binders of Nudt15.
Collapse
Affiliation(s)
- Maciej Lukaszewicz
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland.
| | - Aleksandra Ferenc-Mrozek
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Julia Kokosza
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Anna Stefaniuk
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Janusz Stepinski
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Elzbieta Bojarska
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Edward Darzynkiewicz
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097, Warsaw, Poland
| |
Collapse
|
32
|
Yin J, Wang X, Ge X, Ding F, Shi Z, Ge Z, Huang G, Zhao N, Chen D, Zhang J, Agnihotri S, Cao Y, Ji J, Lin F, Wang Q, Zhou Q, Wang X, You Y, Lu Z, Qian X. Hypoxanthine phosphoribosyl transferase 1 metabolizes temozolomide to activate AMPK for driving chemoresistance of glioblastomas. Nat Commun 2023; 14:5913. [PMID: 37737247 PMCID: PMC10516874 DOI: 10.1038/s41467-023-41663-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Temozolomide (TMZ) is a standard treatment for glioblastoma (GBM) patients. However, TMZ has moderate therapeutic effects due to chemoresistance of GBM cells through less clarified mechanisms. Here, we demonstrate that TMZ-derived 5-aminoimidazole-4-carboxamide (AICA) is converted to AICA ribosyl-5-phosphate (AICAR) in GBM cells. This conversion is catalyzed by hypoxanthine phosphoribosyl transferase 1 (HPRT1), which is highly expressed in human GBMs. As the bona fide activator of AMP-activated protein kinase (AMPK), TMZ-derived AICAR activates AMPK to phosphorylate threonine 52 (T52) of RRM1, the catalytic subunit of ribonucleotide reductase (RNR), leading to RNR activation and increased production of dNTPs to fuel the repairment of TMZ-induced-DNA damage. RRM1 T52A expression, genetic interruption of HPRT1-mediated AICAR production, or administration of 6-mercaptopurine (6-MP), a clinically approved inhibitor of HPRT1, blocks TMZ-induced AMPK activation and sensitizes brain tumor cells to TMZ treatment in mice. In addition, HPRT1 expression levels are positively correlated with poor prognosis in GBM patients who received TMZ treatment. These results uncover a critical bifunctional role of TMZ in GBM treatment that leads to chemoresistance. Our findings underscore the potential of combined administration of clinically available 6-MP to overcome TMZ chemoresistance and improve GBM treatment.
Collapse
Affiliation(s)
- Jianxing Yin
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Gusu School, Nanjing Medical University, 215006, Suzhou, China
| | - Xiefeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
| | - Xin Ge
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, 210029, Nanjing, China
| | - Fangshu Ding
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, 210029, Nanjing, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
| | - Zehe Ge
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, 210029, Nanjing, China
| | - Guang Huang
- Department of Health Inspection and Quarantine, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Ningwei Zhao
- China Exposomics Institute, 200120, Shanghai, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, 210029, Nanjing, China
| | - Dongyin Chen
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
| | - Sameer Agnihotri
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Yuandong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
| | - Fan Lin
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, 211166, Nanjing, China
| | - Qianghu Wang
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Bioinformatics, Nanjing Medical University, 211166, Nanjing, China
| | - Qigang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Xiuxing Wang
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, 211166, Nanjing, China
- National Health Commission Key Laboratory of Antibody Technologies, Nanjing Medical University, 211166, Nanjing, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 310029, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University Cancer Center, Zhejiang University, 310029, Hangzhou, China.
| | - Xu Qian
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, 211166, Nanjing, China.
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, 210029, Nanjing, China.
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
33
|
Kodama M, Toyokawa G, Sugahara O, Sugiyama S, Haratake N, Yamada Y, Wada R, Takamori S, Shimokawa M, Takenaka T, Tagawa T, Kittaka H, Tsuruda T, Tanaka K, Komatsu Y, Nakata K, Imado Y, Yamazaki K, Okamoto I, Oda Y, Takahashi M, Izumi Y, Bamba T, Shimizu H, Yoshizumi T, Nakayama KI. Modulation of host glutamine anabolism enhances the sensitivity of small cell lung cancer to chemotherapy. Cell Rep 2023; 42:112899. [PMID: 37531252 DOI: 10.1016/j.celrep.2023.112899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 06/22/2023] [Accepted: 07/14/2023] [Indexed: 08/04/2023] Open
Abstract
Small cell lung cancer (SCLC) is one of the deadliest human cancers, with a 5-year survival rate of ∼7%. Here, we performed a targeted proteomics analysis of human SCLC samples and thereby identified hypoxanthine phosphoribosyltransferase 1 (HPRT1) in the salvage purine synthesis pathway as a factor that contributes to SCLC malignancy by promoting cell survival in a glutamine-starved environment. Inhibition of HPRT1 by 6-mercaptopurine (6-MP) in combination with methotrexate (MTX), which blocks the de novo purine synthesis pathway, attenuated the growth of SCLC in mouse xenograft models. Moreover, modulation of host glutamine anabolism with the glutamine synthetase inhibitor methionine sulfoximine (MSO) in combination with 6-MP and MTX treatment resulted in marked tumor suppression and prolongation of host survival. Our results thus suggest that modulation of host glutamine anabolism combined with simultaneous inhibition of the de novo and salvage purine synthesis pathways may be of therapeutic benefit for SCLC.
Collapse
Affiliation(s)
- Manabu Kodama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Thoracic Surgery, NHO Kyushu Medical Center, 1-8-1 Jigyohama, Chuo-ku, Fukuoka 810-0065, Japan
| | - Osamu Sugahara
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoki Haratake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Reona Wada
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroki Kittaka
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; LSI Medience Corporation, 1-13-4 Uchikanda, Chiyoda-ku, Tokyo 101-8517, Japan
| | - Takeshi Tsuruda
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yushiro Komatsu
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Keisuke Nakata
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuri Imado
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koji Yamazaki
- Department of Thoracic Surgery, NHO Kyushu Medical Center, 1-8-1 Jigyohama, Chuo-ku, Fukuoka 810-0065, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideyuki Shimizu
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Anticancer Strategies Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
34
|
Alsultan A, Alalwan AA, Alshehri B, Jeraisy MA, Alghamdi J, Alqahtani S, Albassam AA. Interethnic differences in drug response: projected impact of genetic variations in the Saudi population. Pharmacogenomics 2023; 24:685-696. [PMID: 37610881 DOI: 10.2217/pgs-2023-0105] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Ethnicity is known to have an impact on drug responses. This is particularly important for drugs that have a narrow therapeutic window, nonlinearity in pharmacokinetics and are metabolized by enzymes that demonstrate genetic polymorphisms. However, most clinical trials are conducted among Caucasians, which might limit the usefulness of the findings of such studies for other ethnicities. The representation of participants from Saudi Arabia in global clinical trials is low. Therefore, there is a paucity of evidence to assess the impact of ethnic variability in the Saudi population on drug response. In this article, the authors assess the projected impact of genetic polymorphisms in drug-metabolizing enzymes and drug targets on drug response in the Saudi population.
Collapse
Affiliation(s)
- Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A Alalwan
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Bashayer Alshehri
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Majed Al Jeraisy
- Pharmaceutical Care Department, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Jahad Alghamdi
- Saudi Food and Drug Authority, Drug Sector, Riyadh, Saudi Arabia
| | - Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Albassam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
35
|
Yamashita N, Kawahara M, Imai T, Tatsumi G, Asai-Nishishita A, Andoh A. Loss of Nudt15 thiopurine detoxification increases direct DNA damage in hematopoietic stem cells. Sci Rep 2023; 13:11908. [PMID: 37488179 PMCID: PMC10366091 DOI: 10.1038/s41598-023-38952-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
Thiopurines, such as 6-mercaptopurine (6-MP), are widely used as cytotoxic agents and immunosuppressants for leukemia and autoimmune or inflammatory diseases. A nonsynonymous single nucleotide polymorphism (p.Arg139Cys; R139C) of the nucleoside diphosphate-linked moiety X-type motif 15 (NUDT15) gene causes the loss of thiopurine detoxification, inducing myelosuppression. To understand such hematotoxicity, we investigate the effects of NUDT15 R139C on hematopoietic stem cells (HSCs) upon thiopurine administration. Using previously established Nudt15R138C knock-in mice, which mimic myelosuppression in NUDT15R139C homozygous or heterozygous patients following thiopurine administration, we investigated the numerical changes of HSCs and hematopoietic progenitor cells following 6-MP administration using in vivo flowcytometry and ex vivo HSC expansion. Genes differentially expressed between Nudt15+/+ HSCs and Nudt15R138C/R138C HSCs were identified using RNA-sequencing before the emergence of 6-MP-induced HSC-damage. Gene Ontology (GO) and Transcriptional Regulatory Relationships Unraveled by Sentence-based Text Mining (TRRUST) analyses were performed to elucidate the molecular effects of 6-MP on HSCs. In Nudt15R138C/R138C mice, 6-MP induced exhaustion of HSCs faster than that of multipotent progenitors and as fast as that of myeloid-committed progenitors. Ex vivo-expanded Nudt15R138C/R138C HSCs were dose- and time-dependently damaged by 6-MP. GO analysis identified the DNA damage response and cell cycle process as the most strongly influenced processes in Nudt15R138C/R138C HSCs. TRRUST analysis revealed that the Trp53-regulated transcriptional regulatory network is influenced prior to HSC exhaustion in Nudt15R138C/R138C HSCs. The loss of NUDT15 thiopurine detoxification enhances thiopurine-mediated DNA damage via the Trp53 networks in HSCs. Therefore, caution is required in long-term thiopurine use in patients with NUDT15 R139C in view of its adverse effects on HSCs in the form of DNA damage.
Collapse
Affiliation(s)
- Noriaki Yamashita
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Masahiro Kawahara
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan.
| | - Takayuki Imai
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Goichi Tatsumi
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Ai Asai-Nishishita
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Akira Andoh
- Division of Gastroenterology and Hematology, Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
36
|
Flickinger KM, Wilson KM, Rossiter NJ, Hunger AL, Lee TD, Hall MD, Cantor JR. Conditional lethality profiling reveals anticancer mechanisms of action and drug-nutrient interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543621. [PMID: 37333068 PMCID: PMC10274668 DOI: 10.1101/2023.06.04.543621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chemical screening studies have identified drug sensitivities across hundreds of cancer cell lines but most putative therapeutics fail to translate. Discovery and development of drug candidates in models that more accurately reflect nutrient availability in human biofluids may help in addressing this major challenge. Here we performed high-throughput screens in conventional versus Human Plasma-Like Medium (HPLM). Sets of conditional anticancer compounds span phases of clinical development and include non-oncology drugs. Among these, we characterize a unique dual-mechanism of action for brivudine, an agent otherwise approved for antiviral treatment. Using an integrative approach, we find that brivudine affects two independent targets in folate metabolism. We also traced conditional phenotypes for several drugs to the availability of nucleotide salvage pathway substrates and verified others for compounds that seemingly elicit off-target anticancer effects. Our findings establish generalizable strategies for exploiting conditional lethality in HPLM to reveal therapeutic candidates and mechanisms of action.
Collapse
|
37
|
Krancewicz K, Nowicka-Bauer K, Fiedorowicz K, Marciniak B, Taras-Goslinska K. Thiopurines Analogues with Additional Ring: Synthesis, Spectroscopic Properties, and Anticancer Potency. Int J Mol Sci 2023; 24:ijms24108990. [PMID: 37240336 DOI: 10.3390/ijms24108990] [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: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Purine scaffolds constitute a starting point for the synthesis of numerous chemotherapeutics used in treating cancer, viruses, parasites, as well as bacterial and fungal infections. In this work, we synthesized a group of guanosine analogues containing an additional five-membered ring and a sulfur atom at the C-9 position. The spectral, photophysical, and biological properties of the synthesized compounds were investigated. The spectroscopic studies revealed that a combination of the thiocarbonyl chromophore and the tricyclic structure of guanine analogues shifts the absorption region above 350 nm, allowing for selective excitation when present in biological systems. Unfortunately, due to the low fluorescence quantum yield, this process cannot be used to monitor the presence of these compounds in cells. The synthesized compounds were evaluated for their effect on the viability of human cervical carcinoma (HeLa) and mouse fibroblast (NIH/3T3) cells. It was found that all of them display anticancer activity. In vitro studies were preceded by in silico ADME and PASS analyses, which confirmed that the designed compounds are promising candidates for anticancer agents.
Collapse
Affiliation(s)
- Katarzyna Krancewicz
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Karolina Nowicka-Bauer
- Centre for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 10, 61-614 Poznan, Poland
| | - Katarzyna Fiedorowicz
- Nanobiomedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznan, Poland
| | - Bronislaw Marciniak
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
- Centre for Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznanskiego 10, 61-614 Poznan, Poland
| | - Katarzyna Taras-Goslinska
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| |
Collapse
|
38
|
Wu T, Huang S, Feng X, Liu X, James TD, Sun X, Qian X. Visualizing Drug Release from a Stimuli-Responsive Soft Material Based on Amine-Thiol Displacement. ACS APPLIED MATERIALS & INTERFACES 2023; 15:22967-22976. [PMID: 37145981 DOI: 10.1021/acsami.3c02720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In this research, we developed a photoluminescent platform using amine-coupled fluorophores, generated from a single conjugate acceptor containing bis-vinylogous thioesters. Based on the experimental and computational results, the fluorescence turn-on mechanism was proposed to be charge separated induced energy radiative transition for the amine-coupled fluorophore, while the sulfur-containing precursor was not fluorescent since the energy internal conversion occurred through vibrational 2RS- (R represents alkyl groups) as energy acceptor(s). Further utilizing the conjugate acceptor, we establish a new fluorogenic approach via a highly cross-linked soft material to selectively detect cysteine under neutral aqueous conditions. Turn-on fluorescence emission and macroscopic degradation occurred in the presence of cysteine as the stimuli, which can be visually tracked due to the generation of an optical indicator and the cleavage of linkers within the matrix. Furthermore, a novel drug delivery system was constructed, achieving controlled release of sulfhydryl drug (6-mercaptopurine) which was tracked by photoluminescence and high-performance liquid chromatography. The photoluminescent molecules developed herein are suitable for visualizing polymeric degradation, making them suitable for additional "smart" material applications.
Collapse
Affiliation(s)
- Tianhong Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Shiqing Huang
- Singapore University of Technology and Design, 8 Somapah Road, 487372, Singapore
| | - Xing Feng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiaogang Liu
- Singapore University of Technology and Design, 8 Somapah Road, 487372, Singapore
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, P. R. China
| | - Xiaolong Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xuhong Qian
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, P. R. China
| |
Collapse
|
39
|
Teng JY, Yang DP, Tang C, Fang HS, Sun HY, Xiang YN, Li XM, Yang F, Xia RX, Fan F, Liu J, Yu J, Hu JC, Li BS, Li H, Meng FL, Duan CW, Zhou BBS. Targeting DNA polymerase β elicits synthetic lethality with mismatch repair deficiency in acute lymphoblastic leukemia. Leukemia 2023:10.1038/s41375-023-01902-3. [PMID: 37095208 DOI: 10.1038/s41375-023-01902-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 03/26/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
Mismatch repair (MMR) deficiency has been linked to thiopurine resistance and hypermutation in relapsed acute lymphoblastic leukemia (ALL). However, the repair mechanism of thiopurine-induced DNA damage in the absence of MMR remains unclear. Here, we provide evidence that DNA polymerase β (POLB) of base excision repair (BER) pathway plays a critical role in the survival and thiopurine resistance of MMR-deficient ALL cells. In these aggressive resistant ALL cells, POLB depletion and its inhibitor oleanolic acid (OA) treatment result in synthetic lethality with MMR deficiency through increased cellular apurinic/apyrimidinic (AP) sites, DNA strand breaks and apoptosis. POLB depletion increases thiopurine sensitivities of resistant cells, and OA synergizes with thiopurine to kill these cells in ALL cell lines, patient-derived xenograft (PDX) cells and xenograft mouse models. Our findings suggest BER and POLB's roles in the process of repairing thiopurine-induced DNA damage in MMR-deficient ALL cells, and implicate their potentials as therapeutic targets against aggressive ALL progression.
Collapse
Affiliation(s)
- Ji-Yuan Teng
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ding-Peng Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chao Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hou-Shun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Ying Sun
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue-Ning Xiang
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Meng Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui-Xue Xia
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fu Fan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Liu
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiyang Yu
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jin-Chuan Hu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ben-Shang Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fei-Long Meng
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
40
|
Li ZZ, He JY, Wu Q, Liu B, Bu LL. Recent advances in targeting myeloid-derived suppressor cells and their applications to radiotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:233-264. [PMID: 37438019 DOI: 10.1016/bs.ircmb.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogenous immature myeloid cells with potent immune suppressive properties that not only constrain anti-tumor immune activation and functions, promote tumor progression, but also contribute to treatment resistance and tumor relapse. Targeting MDSCs may be a promising new cancer treatment method, but there is still a problem of low treatment efficiency. Combined application with radiotherapy may be a potential method to solve this problem. Drug delivery systems (DDSs) provide more efficient targeted drug delivery capability and can reduce the toxicity and side effects of drugs. Recent advance in DDSs targeting development, recruitment, differentiation, and elimination of MDSCs have shown promising effect in reversing immune inhibition and in overcoming radiotherapy resistance. In this review, we systematically summarized DDSs applied to target MDSCs for the first time, and classified and discussed it according to its different mechanisms of action. In addition, this paper also reviewed the biological characteristics of MDSCs and their role in the initiation, progression, and metastasis of cancer. Moreover, this review also summarizes the role of DDSs targeting MDSCs in radiosensitization. Finally, the future development of DDSs targeting MDSCs is also prospected.
Collapse
Affiliation(s)
- Zi-Zhan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jing-Yu He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
41
|
Liu W, Yasui M, Sassa A, You X, Wan J, Cao Y, Xi J, Zhang X, Honma M, Luan Y. FTO regulates the DNA damage response via effects on cell-cycle progression. MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2023; 887:503608. [PMID: 37003652 DOI: 10.1016/j.mrgentox.2023.503608] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023]
Abstract
The fat mass and obesity-associated protein FTO is an "eraser" of N6-methyladenosine, the most abundant mRNA modification. FTO plays important roles in tumorigenesis. However, its activities have not been fully elucidated and its possible involvement in DNA damage - the early driving event in tumorigenesis - remains poorly characterized. Here, we have investigated the role of FTO in the DNA damage response (DDR) and its underlying mechanisms. We demonstrate that FTO responds to various DNA damage stimuli. FTO is overexpressed in mice following exposure to the promutagens aristolochic acid I and benzo[a]pyrene. Knockout of the FTO gene in TK6 cells, via CRISPR/Cas9, increased genotoxicity induced by DNA damage stimuli (micronucleus and TK mutation assays). Cisplatin- and diepoxybutane-induced micronucleus frequencies and methyl methanesulfonate- and azathioprine-induced TK mutant frequencies were also higher in FTO KO cells. We investigated the potential roles of FTO in DDR. RNA sequencing and enrichment analysis revealed that FTO deletion disrupted the p38 MAPK pathway and inhibited the activation of nucleotide excision repair and cell-cycle-related pathways following cisplatin (DNA intrastrand cross-links) treatment. These effects were confirmed by western blotting and qRT-PCR. FTO deletion impaired cell-cycle arrest at the G2/M phase following cisplatin and diepoxybutane treatment (flow cytometry analysis). Our findings demonstrated that FTO is involved in several aspects of DDR, acting, at least in part, by impairing cell cycle progression.
Collapse
|
42
|
Nguyen TTT, Tanaka Y, Sanada M, Hosaka M, Tamai M, Kagami K, Komatsu C, Somazu S, Harama D, Kasai S, Watanabe A, Akahane K, Goi K, Inukai T. CRISPR/Cas9-Mediated Induction of Relapse-Specific NT5C2 and PRPS1 Mutations Confers Thiopurine Resistance as a Relapsed Lymphoid Leukemia Model. Mol Pharmacol 2023; 103:199-210. [PMID: 36669880 DOI: 10.1124/molpharm.122.000546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
6-Mercaptopurine (6-MP) is a key component in maintenance therapy for childhood acute lymphoblastic leukemia (ALL). Recent next-generation sequencing analysis of childhood ALL clarified the emergence of the relapse-specific mutations of the NT5C2 and PRPS1 genes, which are involved in thiopurine metabolism. In this scenario, minor clones of leukemia cells could acquire the 6-MP-resistant phenotype as a result of the NT5C2 or PRPS1 mutation during chemotherapy (including 6-MP treatment) and confer disease relapse after selective expansion. Thus, to establish new therapeutic modalities overcoming 6-MP resistance in relapsed ALL, human leukemia models with NT5C2 and PRPS1 mutations in the intrinsic genes are urgently required. Here, mimicking the initiation process of the above clinical course, we sought to induce two relapse-specific hotspot mutations (R39Q mutation of the NT5C2 gene and S103N mutation of the PRPS1 gene) into a human lymphoid leukemia cell line by homologous recombination (HR) using the CRISPR/Cas9 system. After 6-MP selection of the cells transfected with Cas9 combined with single-guide RNA and donor DNA templates specific for either of those two mutations, we obtained the sublines with the intended NT5C2-R39Q and PRPS1-S103N mutation as a result of HR. Moreover, diverse in-frame small insertion/deletions were also confirmed in the 6-MP-resistant sublines at the target sites of the NT5C2 and PRPS1 genes as a result of nonhomologous end joining. These sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations in the 6-MP sensitivity and development of therapy overcoming the thiopurine resistance of leukemia cells. SIGNIFICANCE STATEMENT: Mimicking the initiation process of relapse-specific mutations of the NT5C2 and PRPS1 genes in childhood acute lymphoblastic leukemia treated with 6-mercaptopurine (6-MP), this study sought to introduce NT5C2-R39Q and PRPS1-S103N mutations into a human lymphoid leukemia cell line by homologous recombination using the CRISPR/Cas9 system. In the resultant 6-MP-resistant sublines, the intended mutations and diverse in-frame small insertions/deletions were confirmed, indicating that the obtained sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations.
Collapse
Affiliation(s)
- Thao Thu Thi Nguyen
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Yoichi Tanaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masashi Sanada
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masumi Hosaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Minori Tamai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Keiko Kagami
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Chiaki Komatsu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shinpei Somazu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Daisuke Harama
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shin Kasai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Atsushi Watanabe
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Koushi Akahane
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Kumiko Goi
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Takeshi Inukai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| |
Collapse
|
43
|
Gómez-Rubio E, Garcia-Marin J. Molecular dynamics simulations reveal the impact of NUDT15 R139C and R139H variants in structural conformation and dynamics. J Biomol Struct Dyn 2023; 41:14812-14821. [PMID: 36907600 DOI: 10.1080/07391102.2023.2187626] [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: 10/25/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
NUDT15, also known as MTH2, is a member of the NUDIX protein family that catalyzes the hydrolysis of nucleotides and deoxynucleotides, as well as thioguanine analogues. NUDT15 has been reported as a DNA sanitizer in humans, and more recent studies have shown that some genetic variants are related to a poor prognosis in neoplastic and immunologic diseases treated with thioguanine drugs. Despite this, the role of NUDT15 in physiology and molecular biology is quite unclear, as is the mechanism of action of this enzyme. The existence of clinically relevant variants has prompted the study of these enzymes, whose capacity to bind and hydrolyze thioguanine nucleotides is still poorly understood. By using a combination of biomolecular modeling techniques and molecular dynamics, we have studied the monomeric wild type NUDT15 as well as two important variants, R139C and R139H. Our findings reveal not only how nucleotide binding stabilizes the enzyme but also how two loops are responsible for keeping the enzyme in a packed, close conformation. Mutations in α2 helix affect a network of hydrophobic and π-interactions that enclose the active site. This knowledge contributes to the understanding of NUDT15 structural dynamics and will be valuable for the design of new chemical probes and drugs targeting this protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Elena Gómez-Rubio
- Departamento de Biología Estructural y Química, Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Javier Garcia-Marin
- Departamento de Química Orgánica y Química Inorgánica, Instituto de Investigación Química Andrés M. del Río (IQAR), Universidad de Alcalá (IRYCIS), Madrid, Spain
| |
Collapse
|
44
|
Elitzur S, Vora A, Burkhardt B, Inaba H, Attarbaschi A, Baruchel A, Escherich G, Gibson B, Liu HC, Loh M, Moorman AV, Möricke A, Pieters R, Uyttebroeck A, Baird S, Bartram J, Barzilai-Birenboim S, Batra S, Ben-Harosh M, Bertrand Y, Buitenkamp T, Caldwell K, Drut R, Geerlinks AV, Gilad G, Grainger J, Haouy S, Heaney N, Huang M, Ingham D, Krenova Z, Kuhlen M, Lehrnbecher T, Manabe A, Niggli F, Paris C, Revel-Vilk S, Rohrlich P, Sinno MG, Szczepanski T, Tamesberger M, Warrier R, Wolfl M, Nirel R, Izraeli S, Borkhardt A, Schmiegelow K. EBV-driven lymphoid neoplasms associated with pediatric ALL maintenance therapy. Blood 2023; 141:743-755. [PMID: 36332176 DOI: 10.1182/blood.2022016975] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The development of a second malignancy after the diagnosis of childhood acute lymphoblastic leukemia (ALL) is a rare event. Certain second malignancies have been linked with specific elements of leukemia therapy, yet the etiology of most second neoplasms remains obscure and their optimal management strategies are unclear. This is a first comprehensive report of non-Hodgkin lymphomas (NHLs) following pediatric ALL therapy, excluding stem-cell transplantation. We analyzed data of patients who developed NHL following ALL diagnosis and were enrolled in 12 collaborative pediatric ALL trials between 1980-2018. Eighty-five patients developed NHL, with mature B-cell lymphoproliferations as the dominant subtype (56 of 85 cases). Forty-six of these 56 cases (82%) occurred during or within 6 months of maintenance therapy. The majority exhibited histopathological characteristics associated with immunodeficiency (65%), predominantly evidence of Epstein-Barr virus-driven lymphoproliferation. We investigated 66 cases of post-ALL immunodeficiency-associated lymphoid neoplasms, 52 from our study and 14 additional cases from a literature search. With a median follow-up of 4.9 years, the 5-year overall survival for the 66 patients with immunodeficiency-associated lymphoid neoplasms was 67.4% (95% confidence interval [CI], 56-81). Five-year cumulative risks of lymphoid neoplasm- and leukemia-related mortality were 20% (95% CI, 10.2-30) and 12.4% (95% CI, 2.7-22), respectively. Concurrent hemophagocytic lymphohistiocytosis was associated with increased mortality (hazard ratio, 7.32; 95% CI, 1.62-32.98; P = .01). A large proportion of post-ALL lymphoid neoplasms are associated with an immunodeficient state, likely precipitated by ALL maintenance therapy. Awareness of this underrecognized entity and pertinent diagnostic tests are crucial for early diagnosis and optimal therapy.
Collapse
Affiliation(s)
- Sarah Elitzur
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ajay Vora
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Birgit Burkhardt
- Pediatric Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Andre Baruchel
- Department of Pediatric Hematology, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncoogy, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Brenda Gibson
- Department of Paediatric Haematology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Hsi-Che Liu
- Division of Pediatric Hematology/Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Mignon Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital and the Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Centre, Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anja Möricke
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Anne Uyttebroeck
- Department of Paediatric Haematology and Oncology, University Hospital Leuven, Leuven, Leuven, Belgium
| | - Susan Baird
- Department of Haematology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Jack Bartram
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Shlomit Barzilai-Birenboim
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Sandeep Batra
- Pediatric Hematology/Oncology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Miriam Ben-Harosh
- Department of Pediatric Hemato-Oncology, Soroka Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yves Bertrand
- Institut d'Hematologie et d'Oncologie Pediatrique, Hospices Civils de Lyon, Lyon, France
| | - Trudy Buitenkamp
- Amsterdam Academic Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Kenneth Caldwell
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Ricardo Drut
- Department of Pathology, School of Medicine, La Plata National University, La Plata, Argentina
| | | | - Gil Gilad
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - John Grainger
- Faculty of Medical & Human Sciences, University of Manchester and Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie Haouy
- Department of Pediatric Oncology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Nicholas Heaney
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Mary Huang
- Department of Pediatric Hematology Oncology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA
| | - Danielle Ingham
- Paediatric Oncology, Leeds Children's Hospital, Leeds, United Kingdom
| | - Zdenka Krenova
- Department of Pediatric Oncology and Department of Pediatrics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michaela Kuhlen
- Pediatrics and Adolescent Medicine, University of Augsburg, Augsburg, Germany
| | - Thomas Lehrnbecher
- Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Felix Niggli
- Department of Pediatric Oncology, University Children's Hospital, Zurich, Switzerland
| | - Claudia Paris
- Department of Pediatric Oncology and Hematology, Hospital Luis Calvo Mackenna, Santiago, Chile
| | - Shoshana Revel-Vilk
- Shaare Zedek Medical Centre and The Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | | | - Mohamad G Sinno
- Phoenix Children's Hospital, Center for Cancer and Blood Disorders, Phoenix, AZ
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze and Medical University of Silesia, Katowice, Poland
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent Medicine, Kepler University Clinic, Linz, Austria
| | | | - Matthias Wolfl
- Pediatric Oncology, Hematology and Stem Cell Transplantation Program, University Children's Hospital Würzburg, Würzburg, Germany
| | - Ronit Nirel
- Department of Statistics and Data Science, Hebrew University, Jerusalem, Israel
| | - Shai Izraeli
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital, Rigshospitalet, and Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Allele-specific polymerase chain reaction can determine the diplotype of NUDT15 variants in patients with childhood acute lymphoblastic Leukemia. Sci Rep 2023; 13:490. [PMID: 36627439 PMCID: PMC9832159 DOI: 10.1038/s41598-023-27720-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
Mercaptopurine intolerance is an adverse effect of mercaptopurine administration in pediatric patients with acute lymphoblastic leukemia (ALL). NUDT15 variants have emerged as major determinants of mercaptopurine intolerance, especially in the Asian population. Two variants, c.55_56insGAGTCG in exon 1 and c.415C > T in exon 3, were commonly detected in the same allele, named NUDT15*1/*2. Although rare, compound heterozygous mutations also occur, with the two variants on different alleles (NUDT15*3/*6), which may confer tolerance to considerably lesser mercaptopurine dosage. Sanger sequencing or pyrosequencing can determine the NUDT15 variants but not the phase. Here, we designed an allele-specific PCR (AS-PCR) with locked nucleic acid-modified primers. A cohort of 63 patients harboring heterozygous c.55_56insGAGTCG and c.415C > T NUDT15 variations was selected for haplotyping using AS-PCR. Of the 63 patients, 60 harbored the NUDT15*1/*2 variant and three harbored compound heterozygous mutations, including two NUDT15*3/*6 and one NUDT15*2/*7 variants. These findings suggest that AS-PCR can determine NUDT15 diplotype and identify patients with compound heterozygous NUDT15 variants, which may enable precise genetic diagnosis of NUDT15. Nevertheless, a larger clinical trial is required to understand the clinical significance of NUDT15*3/*6 in pediatric patients with ALL because of its low incidence rate and challenges in detecting this variant.
Collapse
|
46
|
Pal S, Sharma A, Mathew SP, Jaganathan BG. Targeting cancer-specific metabolic pathways for developing novel cancer therapeutics. Front Immunol 2022; 13:955476. [PMID: 36618350 PMCID: PMC9815821 DOI: 10.3389/fimmu.2022.955476] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer is a heterogeneous disease characterized by various genetic and phenotypic aberrations. Cancer cells undergo genetic modifications that promote their proliferation, survival, and dissemination as the disease progresses. The unabated proliferation of cancer cells incurs an enormous energy demand that is supplied by metabolic reprogramming. Cancer cells undergo metabolic alterations to provide for increased energy and metabolite requirement; these alterations also help drive the tumor progression. Dysregulation in glucose uptake and increased lactate production via "aerobic glycolysis" were described more than 100 years ago, and since then, the metabolic signature of various cancers has been extensively studied. However, the extensive research in this field has failed to translate into significant therapeutic intervention, except for treating childhood-ALL with amino acid metabolism inhibitor L-asparaginase. Despite the growing understanding of novel metabolic alterations in tumors, the therapeutic targeting of these tumor-specific dysregulations has largely been ineffective in clinical trials. This chapter discusses the major pathways involved in the metabolism of glucose, amino acids, and lipids and highlights the inter-twined nature of metabolic aberrations that promote tumorigenesis in different types of cancer. Finally, we summarise the therapeutic interventions which can be used as a combinational therapy to target metabolic dysregulations that are unique or common in blood, breast, colorectal, lung, and prostate cancer.
Collapse
Affiliation(s)
- Soumik Pal
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Amit Sharma
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Sam Padalumavunkal Mathew
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India,Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, India,*Correspondence: Bithiah Grace Jaganathan,
| |
Collapse
|
47
|
Nazerai L, Willis SC, Yankilevich P, Di Leo L, Bosisio FM, Frias A, Bertolotto C, Nersting J, Thastrup M, Buus S, Thomsen AR, Nielsen M, Rohrberg KS, Schmiegelow K, De Zio D. Thiopurine 6TG treatment increases tumor immunogenicity and response to immune checkpoint blockade. Oncoimmunology 2022; 12:2158610. [PMID: 36545256 PMCID: PMC9762757 DOI: 10.1080/2162402x.2022.2158610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Immune-checkpoint inhibitors (ICI) are highly effective in reinvigorating T cells to attack cancer. Nevertheless, a large subset of patients fails to benefit from ICI, partly due to lack of the cancer neoepitopes necessary to trigger an immune response. In this study, we used the thiopurine 6-thioguanine (6TG) to induce random mutations and thus increase the level of neoepitopes presented by tumor cells. Thiopurines are prodrugs which are converted into thioguanine nucleotides that are incorporated into DNA (DNA-TG), where they can induce mutation through single nucleotide mismatching. In a pre-clinical mouse model of a mutation-low melanoma cell line, we demonstrated that 6TG induced clinical-grade DNA-TG integration resulting in an improved tumor control that was strongly T cell dependent. 6TG exposure increased the tumor mutational burden, without affecting tumor cell proliferation and cell death. Moreover, 6TG treatment re-shaped the tumor microenvironment by increasing T and NK immune cells, making the tumors more responsive to immune-checkpoint blockade. We further validated that 6TG exposure improved tumor control in additional mouse models of melanoma. These findings have paved the way for a phase I/II clinical trial that explores whether treatment with thiopurines can increase the proportion of otherwise treatment-resistant cancer patients who may benefit from ICI therapy (NCT05276284).
Collapse
Affiliation(s)
- Loulieta Nazerai
- Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Shona Caroline Willis
- Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Patricio Yankilevich
- Bioinformatics Core Facility, Instituto de Investigación En Biomedicina de Buenos Aires (Ibioba), Buenos Aires, Argentina
| | - Luca Di Leo
- Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Alex Frias
- Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Corine Bertolotto
- Universite Côte d’Azur, Nice, France,INSERM, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Jacob Nersting
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Maria Thastrup
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Soren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Nielsen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Lyngby, Denmark
| | | | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark,Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark,CONTACT Daniela De Zio Melanoma Research Team, Danish Cancer Society Research Center, Copenhagen, Denmark
| |
Collapse
|
48
|
Zhu X, Chao K, Yang T, Wang XD, Guan S, Tang J, Xie W, Yu AM, Yang QF, Li M, Yang HS, Diao N, Hu PJ, Gao X, Huang M. DNA-Thioguanine Nucleotides as a Marker for Thiopurine Induced Late Leukopenia after Dose Optimizing by NUDT15 C415T in Chinese Patients with IBD. Clin Pharmacol Ther 2022; 112:1236-1242. [PMID: 36002392 DOI: 10.1002/cpt.2730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/15/2022] [Indexed: 01/31/2023]
Abstract
Thiopurine dose optimization by thiopurine-S-methyltransferase (TPMT) or nudix hydrolase-15 (NUDT15) significantly reduced early leucopenia in Asia. However, it fails to avoid the late incidence (> 2 months). Although laboratory monitoring of 6-thioguanine nucleotides (6TGN) to optimize thiopurine dose was suggested in White patients the exact association between leucopenia and 6TGN was controversial in Asian patients. In the present study, we aimed to explore whether DNA-thioguanine nucleotides (DNA-TGs) in leukocytes, compared with 6TGN in erythrocytes, can be a better biomarker for late leucopenia. This was a prospective, observational study. Patients with inflammatory bowel disease (IBD) prescribed thiopurine from February 2019 to December 2019 were recruited. Thiopurine dose was optimized by NUDT15 C415T (rs116855232). DNA-TG and 6TGN levels were determined at the time of late leucopenia or 2 months after the stable dose was obtained. A total of 308 patients were included. Thiopurine induced late leucopenia (white blood cells < 3.5 × 109 /L) were observed in 43 patients (14.0%), who had significantly higher DNA-TG concentration than those without leucopenia (P = 4.1 × 10-9 , 423.3 (~ 342.2 to 565.7) vs. 270.5 (~ 188.1 to 394.3) fmol/μg DNA). No difference in 6TGN concentrations between leucopenia and non-leucopenia was found. With a DNA-TG threshold of 340.1 fmol/μg DNA, 83.7% of leucopenia cases could be identified. Multivariate analysis showed that DNA-TG was an independent risk factor for late leucopenia. Quantification of DNA-TG, rather than 6TGN, can be applied to gauge thiopurine therapy after NUDT15 screening in Chinese patients with IBD.
Collapse
Affiliation(s)
- Xia Zhu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Kang Chao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Ting Yang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xue-Ding Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shaoxing Guan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian Tang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California, USA
| | - Qing Fan Yang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Miao Li
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Hong-Sheng Yang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Na Diao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Pin-Jin Hu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Xiang Gao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Bunea MC, Diculescu VC, Enculescu M, Oprea D, Enache TA. Influence of the Photodegradation of Azathioprine on DNA and Cells. Int J Mol Sci 2022; 23:14438. [PMID: 36430909 PMCID: PMC9692341 DOI: 10.3390/ijms232214438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Azathioprine (AZA) is a pharmacologic immunosuppressive agent administrated in various conditions such as autoimmune disease or to prevent the rejection of organ transplantation. The mechanism of action is based on its biologically active metabolite 6-mercaptopurine (6-MP), which is converted, among others, into thioguanine nucleotides capable of incorporating into replicating DNA, which may act as a strong UV chromophore and trigger DNA oxidation. The interaction between azathioprine and DNA, before and after exposure to solar simulator radiation, was investigated using UV-vis spectrometry and differential pulse voltammetry at a glassy carbon electrode. The results indicated that the interaction of AZA with UV radiation was pH-dependent and occurred with the formation of several metabolites, which induced oxidative damage in DNA, and the formation of DNA-metabolite adducts. Moreover, the viability assays obtained for the L929 cell culture showed that both azathioprine and degraded azathioprine induced a decrease in cell proliferation.
Collapse
Affiliation(s)
| | | | - Monica Enculescu
- National Institute of Materials Physics, Atomistilor 405A, 077125 Magurele, Romania
| | - Daniela Oprea
- National Institute of Materials Physics, Atomistilor 405A, 077125 Magurele, Romania
- Faculty of Physics, University of Bucharest, Atomistilor 405, 077125 Magurele, Romania
| | - Teodor Adrian Enache
- National Institute of Materials Physics, Atomistilor 405A, 077125 Magurele, Romania
| |
Collapse
|
50
|
Krancewicz K, Koput J, Hug GL, Marciniak B, Taras-Goslinska KM. Unusual photophysical properties of a new tricyclic derivative of thiopurines in terms of potential applications. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 281:121620. [PMID: 35853257 DOI: 10.1016/j.saa.2022.121620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
The thio analogues of purine bases have been found to possess notable biological and pharmacological capabilities and have an important role to play as anticancer and immunosuppressive drugs. In this work a new tricyclic analogue of guanosine containing sulfur was synthesized, in particular, DTEG (2',3',5'-tri-O-acetyl-6,9-dithioethanoguanosine). Although there is promise for thiopurine derivatives for biomedical applications, there are some liabilities in regard to their exposure to light. As a preliminary survey for such difficulties with DTEG, this work looks into spectral and photophysical processes of DTEG using time-resolved and steady-state optical excitation. In contrast to other thiopurines, which have long-lived triplets, DTEG is shown to have a short-lived triplet making it less dangerous for singlet-oxygen sensitization. Even in anaerobic solutions, its photoreactivity is negligible. These various unusual photochemical properties of DTEG are consistent with DTEG being very promising as an alternative drug to the currently used 6-thiopurines. DTEG also has some interesting photophysical behavior that is distinct from other thioketones. Although thioketones have an unusual fluorescence violating Kasha's Rule and emitting from the second excited singlet state, DTEG does this also, but, in addition, it shows dual fluorescence by emitting from its first excited singlet as well. The assignments of the nature of these excited states are supported by DFT results. This theory and associated kinetic analysis show quantitatively that the dual fluorescence is, in part, tied to the relatively fast S2 to S1 internal conversion compared to other S2 decays and, in part, tied to the relatively slow nonradiative decay of S1 itself.
Collapse
Affiliation(s)
| | - Jacek Koput
- Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Gordon L Hug
- Radiation Laboratory, University of Notre Dame, Notre Dame 46556, USA
| | - Bronisław Marciniak
- Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznan, Poland; Centre for Advanced Technology, Adam Mickiewicz University, 61-614 Poznan, Poland
| | | |
Collapse
|