1
|
Tisnerat C, Di Ciano S, Pertusati F, Serpi M. Microwave-Accelerated Synthesis of Novel Triphosphate Nucleoside Prodrugs: Expanding the Therapeutic Arsenal of Anticancer Agents. Org Lett 2025; 27:322-327. [PMID: 39689913 PMCID: PMC11731325 DOI: 10.1021/acs.orglett.4c04379] [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: 11/21/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024]
Abstract
In this study, we report for the first time a microwave-accelerated synthesis of purine and pyrimidine nucleoside triphosphate prodrugs, whose γ phosphate is masked with an aryloxy moiety and an amino acid ester (γ-ProTriP). The synthetic utility of this method is illustrated by the synthesis of triphosphate prodrugs of clofarabine and gemcitabine, two FDA-approved anticancer drugs. These new prodrugs showed good chemical and rat serum stability. Remarkably the clofarabine prodrug showed significant cytotoxicity against a panel of cancer cell lines.
Collapse
Affiliation(s)
- Camille Tisnerat
- School
of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, Wales, United Kingdom
| | - Samuele Di Ciano
- School
of Pharmacy and Pharmaceutical Sciences, Redwood Building, King Edwards VII Avenue, CF10 3NB, Cardiff, Wales, United Kingdom
| | - Fabrizio Pertusati
- School
of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, Wales, United Kingdom
| | - Michaela Serpi
- School
of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, Wales, United Kingdom
| |
Collapse
|
2
|
Fang SS, Hu X, Li MH, Qi S, Xie T, Wang JB, Yao HQ, Zhang J, Zhang JH, Zhu L, Shang M. Ligand-Enabled Cu-Catalyzed Stereoselective Synthesis of P-Stereogenic ProTides. J Am Chem Soc 2024; 146:31339-31347. [PMID: 39496113 DOI: 10.1021/jacs.4c12920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Nucleoside analogues have seen significant advancements in treating viral infections and cancer through ProTide technology, leading to a series of FDA-approved drugs such as sofosbuvir, tenofovir alafenamide, and remdesivir. The stereochemical configuration at the phosphorus center of ProTides significantly influences their pharmacological properties, necessitating efficient stereoselective synthesis. Traditional methods using chiral auxiliaries or nonracemic phosphorylating agents are labor-intensive and inefficient, while recent organocatalytic approaches, despite their promise, still face limitations. Herein, we present a novel approach employing chiral metal complexes for the stereoselective assembly of P-stereogenic ProTides via asymmetric P-O bond formation. This approach leverages a chiral metal catalyst to activate the electrophilic phosphorylating reagent, facilitating a base-promoted nucleophilic replacement pathway. Our protocol, featuring mild reaction conditions and broad applicability, enables the highly stereoselective synthesis of previously inaccessible (S,RP) and (R,SP)-ProTide derivatives. The practical utility of this method is demonstrated through the preparation of pharmaceutically relevant ProTide targets and mechanistic studies were conducted to elucidate the reaction pathway, offering significant advancements for drug development and pharmaceutical research.
Collapse
Affiliation(s)
- Shuai-Shuai Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - XiangJun Hu
- Department of Rehabilitation Medicine, Shanghai Institute of Rehabilitation with Integrated Western and Chinese Traditional Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ming-Hong Li
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
- Xiangfu Laboratory, Jiashan 314102, China
| | - Shuang Qi
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200217, China
| | - Tian Xie
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jia-Bao Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
- Xiangfu Laboratory, Jiashan 314102, China
| | - Hong-Qing Yao
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian Zhang
- Department of Rehabilitation Medicine, Shanghai Institute of Rehabilitation with Integrated Western and Chinese Traditional Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun-Hua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200217, China
| | - Ming Shang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
- Xiangfu Laboratory, Jiashan 314102, China
| |
Collapse
|
3
|
Lawrence S, Lin J, Khurshid A, Utami W, Singhania R, Ashraf S, Thorn GJ, Mangangcha IR, Spriggs K, Kim DH, Barrett D, de Moor CH. Cordycepin generally inhibits growth factor signal transduction in a systems pharmacology study. FEBS Lett 2024. [PMID: 39508147 DOI: 10.1002/1873-3468.15046] [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: 05/31/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024]
Abstract
Cordycepin (3' deoxyadenosine) has been widely researched as a potential cancer therapy, but many diverse mechanisms of action have been proposed. Here, we confirm that cordycepin triphosphate is likely to be the active metabolite of cordycepin and that it consistently represses growth factor-induced gene expression. Bioinformatic analysis, quantitative PCR and western blotting confirmed that cordycepin blocks the PI3K/AKT/mTOR and/or MEK/ERK pathways in six cell lines and that AMPK activation is not required. The effects of cordycepin on translation through mTOR pathway repression were detectable within 30 min, indicating a rapid process. These data therefore indicate that cordycepin has a universal mechanism of action, acting as cordycepin triphosphate on an as yet unknown target molecule involved in growth factor signalling.
Collapse
Affiliation(s)
- Steven Lawrence
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Jialiang Lin
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Asma Khurshid
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Wahyu Utami
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Richa Singhania
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Sadaf Ashraf
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Graeme J Thorn
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | | | - Keith Spriggs
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Dong-Hyun Kim
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - David Barrett
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| | - Cornelia H de Moor
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, UK
| |
Collapse
|
4
|
Zhang SY, Fu GN, Du LH, Lin H, Zhang AY, Xie HJ, Sheng ZK, Xue MM, Yan BL, Liu Y, Ruan ZX, Pan BL, Zhou TY, Luo XP. Continuous flow biocatalysis: synthesis of purine nucleoside esters catalyzed by lipase TL IM from Thermomyces lanuginosus. RSC Adv 2024; 14:10953-10961. [PMID: 38577433 PMCID: PMC10993233 DOI: 10.1039/d4ra00097h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Purine nucleoside ester is one of the derivatives of purine nucleoside, which has antiviral and anticancer activities. In this work, a continuous flow synthesis of purine nucleoside esters catalyzed by lipase TL IM from Thermomyces lanuginosus was successfully achieved. Various parameters including solvent, reaction temperature, reaction time/flow rate and substrate ratio were investigated. The best yields were obtained with a continuous flow microreactor for 35 min at 50 °C with the substrate ratio of 1 : 5 (nucleosides to vinyl esters) in the solvent of tert-amyl alcohol. 12 products were efficiently synthesized with yields of 78-93%. Here we reported for the first time the use of lipase TL IM from Thermomyces lanuginosus in the synthesis of purine nucleoside esters. The significant advantages of this methodology are a green solvent and mild conditions, a simple work-up procedure and the highly reusable biocatalyst. This research provides a new technique for rapid synthesis of anticancer and antiviral nucleoside drugs and is helpful for further screening of drug activity.
Collapse
Affiliation(s)
- Shi-Yi Zhang
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Guo-Neng Fu
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Li-Hua Du
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Hang Lin
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Ao-Ying Zhang
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Han-Jia Xie
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Zhi-Kai Sheng
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Miao-Miao Xue
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Bing-Lin Yan
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Yi Liu
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Zhi-Xuan Ruan
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Bing-Le Pan
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Tong-Yao Zhou
- College of Pharmaceutical Science, ZheJiang University of Technology Hangzhou 310014 China +86-189-690-693-99
| | - Xi-Ping Luo
- Zhejiang Provincial Key Laboratory of Chemical Utilization of Forestry Biomass, Zhejiang A&F University Hangzhou 311300 China
| |
Collapse
|
5
|
Giurini EF, Godla A, Gupta KH. Redefining bioactive small molecules from microbial metabolites as revolutionary anticancer agents. Cancer Gene Ther 2024; 31:187-206. [PMID: 38200347 PMCID: PMC10874892 DOI: 10.1038/s41417-023-00715-x] [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: 08/21/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024]
Abstract
Cancer treatment remains a significant challenge due to issues such as acquired resistance to conventional therapies and the occurrence of adverse treatment-related toxicities. In recent years, researchers have turned their attention to the microbial world in search of novel and effective drugs to combat this devastating disease. Microbial derived secondary metabolites have proven to be a valuable source of biologically active compounds, which exhibit diverse functions and have demonstrated potential as treatments for various human diseases. The exploration of these compounds has provided valuable insights into their mechanisms of action against cancer cells. In-depth studies have been conducted on clinically established microbial metabolites, unraveling their anticancer properties, and shedding light on their therapeutic potential. This review aims to comprehensively examine the anticancer mechanisms of these established microbial metabolites. Additionally, it highlights the emerging therapies derived from these metabolites, offering a glimpse into the immense potential they hold for anticancer drug discovery. Furthermore, this review delves into approved treatments and major drug candidates currently undergoing clinical trials, focusing on specific molecular targets. It also addresses the challenges and issues encountered in the field of anticancer drug research and development. It also presents a comprehensive exposition of the contemporary panorama concerning microbial metabolites serving as a reservoir for anticancer agents, thereby illuminating their auspicious prospects and the prospect of forthcoming strides in the domain of cancer therapeutics.
Collapse
Affiliation(s)
- Eileena F Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Aishvarya Godla
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Kajal H Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL, 60612, USA.
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
6
|
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
|
7
|
Jiang L, Pan T, Lv Q, Yuan W, Liu X, Qu X, Luo D, Wan S, Cui S. Novel ProTide prodrugs of 5-fluoro-2'-deoxyuridine for the treatment of liver cancer. Eur J Med Chem 2023; 260:115763. [PMID: 37659196 DOI: 10.1016/j.ejmech.2023.115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/08/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
ProTide prodrug technology has emerged as a promising way for the development of anti-viral and anti-tumor drugs, whereas, there are fewer applications for the treatment of liver cancer. Herein, a series of distinct 3'-ester ProTide prodrugs of 5-fluoro-2'-deoxyuridine (FdUR) were synthesized and evaluated for their anti-liver cancer activity. The most efficient prodrug 11b reached a sub-micromolar activity (IC50 = 0.42 ± 0.13 μM) against HepG2 and over 100-fold and 200-fold improvements compared to 5-FU, respectively. 11b also demonstrated favorable selectivity towards normal liver cells L-02 (IC50 > 100 μM). In vitro metabolic stability studies revealed that 11b is stable in the plasma and could be activated rapidly in the liver, which supported that 11b is liver-targeted. Importantly, to more accurately evaluate the anti-HCC activity of 11b, the liver orthotopic model was built and 11b significantly suppressed tumor growth (TGI = 75.5%) at a dose of 60 mg/kg/2d in vivo without obvious toxicity. Overall, these promising results indicated that 11b could serve as a safe and effective prodrug of 5-FU nucleoside for liver cancer therapy.
Collapse
Affiliation(s)
- Leilei Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Ting Pan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Qin Lv
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Wenmin Yuan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Xiaochun Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dongdong Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China.
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China.
| | - Shuxiang Cui
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
8
|
Chen L, Zheng X, Huang H, Feng C, Wu S, Chen R, Jiang H, Yuan M, Fu Y, Ying H, Zhou J, Jiang J. Cordycepin synergizes with CTLA-4 blockade to remodel the tumor microenvironment for enhanced cancer immunotherapy. Int Immunopharmacol 2023; 124:110786. [PMID: 37611443 DOI: 10.1016/j.intimp.2023.110786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
The strategy of using immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, leading to remarkable clinical outcomes. However, certain cancer types and patient demographics continue to face unique challenges. As a result, it is vital to investigate combination therapies that involve ICIs to boost therapeutic efficacy. Cordycepin, an adenosine derivative composed of adenine and pentose, holds immense promise for treating inflammation and cancer. Our recent research has demonstrated that the combined treatment of cordycepin and the anti-CD47 antibody significantly curtails tumor growth and extends the lifespan of tumor-bearing mice. In the current study, we showed that the combination of cordycepin and CTLA-4 blockade had a profound impact on suppressing tumor growth. We utilized the MC38 and CT26 tumor models to evaluate the therapeutic effect of cordycepin, CTLA-4 blockade, and their combined approach. Flow cytometry results unveiled that cordycepin, when combined with CTLA-4 blockade, considerably augmented the presence of tumor-infiltrating CD8+T cells and diminished the population of Foxp3+Tregs within the tumor microenvironment (TME). Additionally, we employed single-cell analysis to examine the TME's reconfiguration upon the combined treatment of anti-CTLA-4 and cordycepin. We observed a significant impact on inhibiting tumor growth and substantially extended survival in tumor-bearing mice. Our data also demonstrated an increased proportion of effector CD8+T cells in the combined treatment group compared to all other groups, while exhausted CD8+T cells diminished in the combined group compared to the anti-CTLA-4 treatment alone. In conclusion, our findings supported the idea that combining cordycepin and CTLA-4 blockade could modify the effector and exhaustion status of CD8+T cells, thereby bolstering CD8+T-cell-mediated anti-tumor immunity in the TME. Collectively, our current study successfully established a combination therapeutic strategy utilizing cordycepin and CTLA-4 blockade. This strategy demonstrated a significant synergistic effect against cancer, highlighting its importance in cancer treatment.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Chen Feng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Rongzhang Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Hongwei Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Maoling Yuan
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China
| | - Yuanyuan Fu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Department of Gynecology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, China
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, China
| | - Jun Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Jiangsu, Changzhou 213003, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
9
|
Priya PS, Murugan R, Almutairi BO, Arokiyaraj S, Shanjeev P, Arockiaraj J. Delineating the protective action of cordycepin against cadmium induced oxidative stress and gut inflammation through downregulation of NF-κB pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104246. [PMID: 37595934 DOI: 10.1016/j.etap.2023.104246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/03/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023]
Abstract
Cadmium (Cd) exposure is known to cause gut inflammation. In this study, we investigated the protective effects of cordycepin, a natural compound with pharmacological properties, against gut inflammation induced by Cd exposure. Using zebrafish larvae and colon cell line models, we examined the impact of cordycepin on Cd-induced toxicity and inflammation. Zebrafish larvae were exposed to Cd (2 µg/mL) and treated with different concentrations of cordycepin (12.5, 25 and 50 µg/mL). Cordycepin treatment significantly reduced Cd-induced embryotoxicity in zebrafish larvae. It also alleviated Cd-induced oxidative stress by reducing reactive oxygen species (ROS), lipid peroxidation and apoptosis. Furthermore, cordycepin treatment normalized the levels of liver-related biomarkers affected due to Cd exposure. Additionally, cordycepin (50 µg/mL) demonstrated a significant reduction in Cd bioaccumulation and downregulated the expression of inflammatory genes in both zebrafish larval gut and colon cell lines. These findings suggest that cordycepin could be an effective agent against Cd-induced gut inflammation.
Collapse
Affiliation(s)
- P Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur 603203, Tamil Nadu, India
| | - Raghul Murugan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur 603203, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - P Shanjeev
- SG's Supreme Organics, Plot 148, Sri Valli Nagar, Nandhivaram Village, Guduvancherry, Chennai 603202, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur 603203, Tamil Nadu, India.
| |
Collapse
|
10
|
Prescott TAK, Hill R, Mas-Claret E, Gaya E, Burns E. Fungal Drug Discovery for Chronic Disease: History, New Discoveries and New Approaches. Biomolecules 2023; 13:986. [PMID: 37371566 DOI: 10.3390/biom13060986] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Fungal-derived drugs include some of the most important medicines ever discovered, and have proved pivotal in treating chronic diseases. Not only have they saved millions of lives, but they have in some cases changed perceptions of what is medically possible. However, now the low-hanging fruit have been discovered it has become much harder to make the kind of discoveries that have characterised past eras of fungal drug discovery. This may be about to change with new commercial players entering the market aiming to apply novel genomic tools to streamline the discovery process. This review examines the discovery history of approved fungal-derived drugs, and those currently in clinical trials for chronic diseases. For key molecules, we discuss their possible ecological functions in nature and how this relates to their use in human medicine. We show how the conservation of drug receptors between fungi and humans means that metabolites intended to inhibit competitor fungi often interact with human drug receptors, sometimes with unintended benefits. We also plot the distribution of drugs, antimicrobial compounds and psychoactive mushrooms onto a fungal tree and compare their distribution to those of all fungal metabolites. Finally, we examine the phenomenon of self-resistance and how this can be used to help predict metabolite mechanism of action and aid the drug discovery process.
Collapse
Affiliation(s)
| | - Rowena Hill
- Earlham Institute, Norwich NR4 7UZ, Norfolk, UK
| | | | - Ester Gaya
- Royal Botanic Gardens, Kew, Richmond TW9 3AB, Surrey, UK
| | - Edie Burns
- Royal Botanic Gardens, Kew, Richmond TW9 3AB, Surrey, UK
| |
Collapse
|
11
|
Tan H, Wang L, Wang H, Cheng Y, Li X, Wan H, Liu C, Liu T, Li Q. Engineering Komagataella phaffii to biosynthesize cordycepin from methanol which drives global metabolic alterations at the transcription level. Synth Syst Biotechnol 2023; 8:242-252. [PMID: 37007278 PMCID: PMC10060148 DOI: 10.1016/j.synbio.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Cordycepin has the potential to be an alternative to the disputed herbicide glyphosate. However, current laborious and time-consuming production strategies at low yields based on Cordyceps militaris lead to extremely high cost and restrict its application in the field of agriculture. In this study, Komagataella phaffii (syn. Pichia pastoris) was engineered to biosynthesize cordycepin from methanol, which could be converted from CO2. Combined with fermentation optimization, cordycepin content in broth reached as high as 2.68 ± 0.04 g/L within 168 h, around 15.95 mg/(L·h) in productivity. Additionally, a deaminated product of cordycepin was identified at neutral or weakly alkaline starting pH during fermentation. Transcriptome analysis found the yeast producing cordycepin was experiencing severe inhibition in methanol assimilation and peroxisome biogenesis, responsible for delayed growth and decreased carbon flux to pentose phosphate pathway (PPP) which led to lack of precursor supply. Amino acid interconversion and disruption in RNA metabolism were also due to accumulation of cordycepin. The study provided a unique platform for the manufacture of cordycepin based on the emerging non-conventional yeast and gave practical strategies for further optimization of the microbial cell factory.
Collapse
Affiliation(s)
- Huiping Tan
- Department of Life and Health, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, China
| | - Liang Wang
- School of Biological Engineering, Dalian Polytechnic University, No. 1 Qinggongyuan Road, Ganjingzi District, Dalian, 116034, China
| | - Huiguo Wang
- Department of Life and Health, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, China
| | - Yanghao Cheng
- Department of Life and Health, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, China
| | - Xiang Li
- Department of Life and Health, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, China
| | - Huihui Wan
- Analytical Instrumentation Centre, Dalian University of Technology, No.2 Linggong Road, Dalian, 116024, China
| | - Chenguang Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Tian Liu
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, 116024, China
| | - Qian Li
- Department of Life and Health, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, China
| |
Collapse
|
12
|
Zhang L, Qi K, Xu J, Xing Y, Wang X, Tong L, He Z, Xu W, Li X, Jiang Y. Design, Synthesis, and Anti-Cancer Evaluation of Novel Cyclic Phosphate Prodrug of Gemcitabine. J Med Chem 2023; 66:4150-4166. [PMID: 36867101 DOI: 10.1021/acs.jmedchem.3c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
ProTide and cyclic phosphate ester are two successful prodrug technologies to overcome the limitations of nucleoside drugs, among which the cyclic phosphate ester strategy has not been widely used in the optimization of gemcitabine. Herein, we designed a series of novel ProTide and cyclic phosphate ester prodrugs of gemcitabine. Cyclic phosphate ester derivative 18c exhibits much higher anti-proliferative activity than positive control NUC-1031 with IC50s of 3.6-19.2 nM on multiple cancer cells. The metabolic pathway of 18c demonstrates that 18c's bioactive metabolites prolong its anti-tumor activity. More importantly, we separated the two P chiral diastereomers of gemcitabine cyclic phosphate ester prodrugs for the first time, revealing their similar cytotoxic potency and metabolic profile. 18c displays significant in vivo anti-tumor activity in both 22Rv1 and BxPC-3 xenograft tumor models. These results suggest that compound 18c is a promising anti-tumor candidate for treating human castration-resistant prostate and pancreatic cancer.
Collapse
Affiliation(s)
- Liang Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, P.R. China
| | - Kangjing Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Xu
- Oncology and Immunology Unit, Research Service Division, WuXi AppTec, Nantong 226299, China
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yan Xing
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, P.R. China
| | - Xuejian Wang
- College of Pharmacy, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Ling Tong
- Oncology and Immunology Unit, Research Service Division, WuXi AppTec, Nantong 226299, China
| | - Zengguo He
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Wenfang Xu
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, P.R. China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, P.R. China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, P.R. China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, P.R. China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
13
|
Yan VC, Barekatain Y, Lin YH, Satani N, Hammoudi N, Arthur K, Georgiou DK, Jiang Y, Sun Y, Marszalek JR, Millward SW, Muller FL. Comparative Pharmacology of a Bis-Pivaloyloxymethyl Phosphonate Prodrug Inhibitor of Enolase after Oral and Parenteral Administration. ACS Pharmacol Transl Sci 2023; 6:245-252. [PMID: 36798479 PMCID: PMC9926520 DOI: 10.1021/acsptsci.2c00216] [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: 11/05/2022] [Indexed: 01/08/2023]
Abstract
Metabolically labile prodrugs can experience stark differences in catabolism incurred by the chosen route of administration. This is especially true for phosph(on)ate prodrugs, in which successive promoiety removal transforms a lipophilic molecule into increasingly polar compounds. We previously described a phosphonate inhibitor of enolase (HEX) and its bis-pivaloyloxymethyl ester prodrug (POMHEX) capable of eliciting strong tumor regression in a murine model of enolase 1 (ENO1)-deleted glioblastoma following parenteral administration. Here, we characterize the pharmacokinetics and pharmacodynamics of these enolase inhibitors in vitro and in vivo after oral and parenteral administration. In support of the historical function of lipophilic prodrugs, the bis-POM prodrug significantly improves cell permeability of and rapid hydrolysis to the parent phosphonate, resulting in rapid intracellular loading of peripheral blood mononuclear cells in vitro and in vivo. We observe the influence of intracellular trapping in vivo on divergent pharmacokinetic profiles of POMHEX and its metabolites after oral and parenteral administration. This is a clear demonstration of the tissue reservoir effect hypothesized to explain phosph(on)ate prodrug pharmacokinetics but has heretofore not been explicitly demonstrated.
Collapse
Affiliation(s)
- Victoria C. Yan
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Yasaman Barekatain
- Department
of Cancer Biology, University of Texas MD
Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Yu-Hsi Lin
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Nikunj Satani
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Naima Hammoudi
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Kenisha Arthur
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Dimitra K. Georgiou
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Yongying Jiang
- Institute
of Applied Cancer Science, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Yuting Sun
- Institute
of Applied Cancer Science, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Joseph R. Marszalek
- Center
for Co-Clinical Trials, University of Texas
MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Steven W. Millward
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| | - Florian L. Muller
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77030-4000, United States
| |
Collapse
|
14
|
Roy B, Navarro V, Peyrottes S. Prodrugs of Nucleoside 5'-Monophosphate Analogues: Overview of the Recent Literature Concerning their Synthesis and Applications. Curr Med Chem 2023; 30:1256-1303. [PMID: 36093825 DOI: 10.2174/0929867329666220909122820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Nucleoside analogues are widely used as anti-infectious and antitumoral agents. However, their clinical use may face limitations associated with their physicochemical properties, pharmacokinetic parameters, and/or their peculiar mechanisms of action. Indeed, once inside the cells, nucleoside analogues require to be metabolized into their corresponding (poly-)phosphorylated derivatives, mediated by cellular and/or viral kinases, in order to interfere with nucleic acid biosynthesis. Within this activation process, the first-phosphorylation step is often the limiting one and to overcome this limitation, numerous prodrug approaches have been proposed. Herein, we will focus on recent literature data (from 2015 and onwards) related to new prodrug strategies, the development of original synthetic approaches and novel applications of nucleotide prodrugs (namely pronucleotides) leading to the intracellular delivery of 5'-monophosphate nucleoside analogues.
Collapse
Affiliation(s)
- Béatrice Roy
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| | - Valentin Navarro
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| | - Suzanne Peyrottes
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| |
Collapse
|
15
|
Shahid AM, Um IH, Elshani M, Zhang Y, Harrison DJ. NUC-7738 regulates β-catenin signalling resulting in reduced proliferation and self-renewal of AML cells. PLoS One 2022; 17:e0278209. [PMID: 36520954 PMCID: PMC9754587 DOI: 10.1371/journal.pone.0278209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/13/2022] [Indexed: 12/23/2022] Open
Abstract
Acute myeloid leukemia (AML) stem cells are required for the initiation and maintenance of the disease. Activation of the Wnt/β-catenin pathway is required for the survival and development of AML leukaemia stem cells (LSCs) and therefore, targeting β-catenin is a potential therapeutic strategy. NUC-7738, a phosphoramidate transformation of 3'-deoxyadenosine (3'-dA) monophosphate, is specifically designed to generate the active anti-cancer metabolite 3'-deoxyadenosine triphosphate (3'-dATP) intracellularly, bypassing key limitations of breakdown, transport, and activation. NUC-7738 is currently in a Phase I/II clinical study for the treatment of patients with advanced solid tumors. Protein expression and immunophenotypic profiling revealed that NUC-7738 caused apoptosis in AML cell lines through reducing PI3K-p110α, phosphorylated Akt (Ser473) and phosphorylated GSK3β (Ser9) resulting in reduced β-catenin, c-Myc and CD44 expression. NUC-7738 reduced β-catenin nuclear expression in AML cells. NUC-7738 also decreased the percentage of CD34+ CD38- CD123+ (LSC-like cells) from 81% to 47% and reduced the total number and size of leukemic colonies. These results indicate that therapeutic targeting of the PI3K/Akt/GSK3β axis can inhibit β-catenin signalling, resulting in reduced clonogenicity and eventual apoptosis of AML cells.
Collapse
Affiliation(s)
| | - In Hwa Um
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Mustafa Elshani
- School of Medicine, University of St Andrews, St Andrews, United Kingdom,NuCana plc, Edinburgh, United Kingdom
| | - Ying Zhang
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - David James Harrison
- School of Medicine, University of St Andrews, St Andrews, United Kingdom,NuCana plc, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Serpi M, Ferrari V, McGuigan C, Ghazaly E, Pepper C. Synthesis and Characterization of NUC-7738, an Aryloxy Phosphoramidate of 3'-Deoxyadenosine, as a Potential Anticancer Agent. J Med Chem 2022; 65:15789-15804. [PMID: 36417756 PMCID: PMC9743095 DOI: 10.1021/acs.jmedchem.2c01348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Indexed: 11/24/2022]
Abstract
3'-Deoxyadenosine (3'-dA, Cordycepin, 1) is a nucleoside analogue with anticancer properties, but its clinical development has been hampered due to its deactivation by adenosine deaminase (ADA) and poor cellular uptake due to low expression of the human equilibrative transporter (hENT1). Here, we describe the synthesis and characterization of NUC-7738 (7a), a 5'-aryloxy phosphoramidate prodrug of 3'-dA. We show in vitro evidence that 7a is an effective anticancer drug in a panel of solid and hematological cancer cell lines, showing its preferential cytotoxic effects on leukemic stem cells. We found that unlike 3'-dA, the activity of 7a was independent of hENT1 and kinase activity. Furthermore, it was resistant to ADA metabolic deactivation. Consistent with these findings, 7a showed increased levels of intracellular 3'-deoxyadenosine triphosphate (3'-dATP), the active metabolite. Mechanistically, levels of intracellular 3'-dATP were strongly associated with in vitro potency. NUC-7738 is now in Phase II, dose-escalation study in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Michaela Serpi
- School
of Chemistry, Cardiff University Main Building, Park Place, Cardiff CF10 3AT, Wales, U.K.
| | - Valentina Ferrari
- School
of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, King Edward VII Avenue, Cardiff CF10 3NB, U.K.
| | - Christopher McGuigan
- School
of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, King Edward VII Avenue, Cardiff CF10 3NB, U.K.
| | - Essam Ghazaly
- Centre
for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, U.K.
| | - Chris Pepper
- Brighton
and Sussex Medical School, University of
Sussex, Brighton BN1 9PX, U.K.
| |
Collapse
|
17
|
Vatandaslar H. A Systematic Study on the Optimal Nucleotide Analogue Concentration and Rate Limiting Nucleotide of the SARS-CoV-2 RNA-Dependent RNA Polymerase. Int J Mol Sci 2022; 23:ijms23158302. [PMID: 35955442 PMCID: PMC9369030 DOI: 10.3390/ijms23158302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/23/2022] [Accepted: 07/24/2022] [Indexed: 02/01/2023] Open
Abstract
The current COVID-19 pandemic has highlighted the necessity of more efficient antiviral compounds. The antiviral efficacy of adenosine-based analogs, the main repurposed drugs for SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) inhibition, is mainly assessed through in vitro or cell-free polymerization assays, under arbitrary conditions that do not reflect the physiological environment. We show that SARS-CoV-2 RdRp inhibition efficiency of remdesivir and cordycepin, two common adenosine analogs, is influenced by endogenous adenosine level, and that the current clinically approved concentrations for COVID-19 treatment are suboptimal for effective RdRp inhibition. Furthermore, we identified GTP as the rate-limiting nucleotide of SARS-CoV-2 replication. Our results demonstrate that nucleotide sensitivity of the RdRp complex and competition of nucleoside analog drugs against endogenous concentrations of nucleotides are crucial elements to be considered when designing new SARS-CoV-2 antiviral compounds.
Collapse
Affiliation(s)
- Hasan Vatandaslar
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
18
|
Cordycepin (3′-Deoxyadenosine) Suppresses Heat Shock Protein 90 Function and Targets Tumor Growth in an Adenosine Deaminase-Dependent Manner. Cancers (Basel) 2022; 14:cancers14133122. [PMID: 35804893 PMCID: PMC9264932 DOI: 10.3390/cancers14133122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Alterations in metabolism and energy production are increasingly being recognized as important drivers of neoplasia, raising the possibility that metabolic analogs could disrupt oncogenic pathways. 3′-deoxyadenosine, also known as cordycepin, is an adenosine analog that inhibits the growth of several types of cancer. However, the effects of cordycepin have only been examined in a limited number of tumor types, and its mechanism of action is poorly understood. We found that cordycepin slows the growth and promotes apoptosis in uveal melanoma, as well as a range of other hard-to-treat malignancies, including retinoblastoma, atypical teratoid rhabdoid tumors, and diffuse midline gliomas. Interestingly, these effects were dependent on low adenosine deaminase (ADA) expression or activity. Inhibition of ADA using either siRNA or pharmacologic approaches sensitized tumors with higher ADA to cordycepin in vitro and in vivo, with increased apoptosis, reduced clonogenic capacity, and slower migration of neoplastic cells. Our studies suggest that ADA is both a biomarker predicting response to cordycepin and a target for combination therapy. We also describe a novel mechanism of action for cordycepin: competition with adenosine triphosphate (ATP) in binding to Hsp90, resulting in impaired processing of oncogenic Hsp90 client proteins.
Collapse
|
19
|
Kontogiannatos D, Koutrotsios G, Xekalaki S, Zervakis GI. Biomass and Cordycepin Production by the Medicinal Mushroom Cordyceps militaris-A Review of Various Aspects and Recent Trends towards the Exploitation of a Valuable Fungus. J Fungi (Basel) 2021; 7:jof7110986. [PMID: 34829273 PMCID: PMC8621325 DOI: 10.3390/jof7110986] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Cordyceps militaris is an entomopathogenic ascomycete with similar pharmacological importance to that of the wild caterpillar fungus Ophiocordyceps sinensis. C. militaris has attracted significant research and commercial interest due to its content in bioactive compounds beneficial to human health and the relative ease of cultivation under laboratory conditions. However, room for improvement exists in the commercial-scale cultivation of C. militaris and concerns issues principally related to appropriate strain selection, genetic degeneration of cultures, and substrate optimization. In particular, culture degeneration-usually expressed by abnormal fruit body formation and reduced sporulation-results in important economic losses and is holding back investors and potential growers (mainly in Western countries) from further developing this highly promising sector. In the present review, the main factors that influence the generation of biomass and metabolites (with emphasis on cordycepin biosynthesis) by C. militaris are presented and evaluated in conjunction with the use of a wide range of supplements or additives towards the enhancement of fungal productivity in large-scale cultivation processes. Moreover, physiological and genetic factors that increase or reduce the manifestation of strain degeneration in C. militaris are outlined. Finally, methodologies for developing protocols to be used in C. militaris functional biology studies are discussed.
Collapse
|