1
|
Xu R, He X, Xu J, Yu G, Wu Y. Immunometabolism: signaling pathways, homeostasis, and therapeutic targets. MedComm (Beijing) 2024; 5:e789. [PMID: 39492834 PMCID: PMC11531657 DOI: 10.1002/mco2.789] [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: 10/19/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Immunometabolism plays a central role in sustaining immune system functionality and preserving physiological homeostasis within the organism. During the differentiation and activation, immune cells undergo metabolic reprogramming mediated by complex signaling pathways. Immune cells maintain homeostasis and are influenced by metabolic microenvironmental cues. A series of immunometabolic enzymes modulate immune cell function by metabolizing nutrients and accumulating metabolic products. These enzymes reverse immune cells' differentiation, disrupt intracellular signaling pathways, and regulate immune responses, thereby influencing disease progression. The huge population of immune metabolic enzymes, the ubiquity, and the complexity of metabolic regulation have kept the immune metabolic mechanisms related to many diseases from being discovered, and what has been revealed so far is only the tip of the iceberg. This review comprehensively summarized the immune metabolic enzymes' role in multiple immune cells such as T cells, macrophages, natural killer cells, and dendritic cells. By classifying and dissecting the immunometabolism mechanisms and the implications in diseases, summarizing and analyzing advancements in research and clinical applications of the inhibitors targeting these enzymes, this review is intended to provide a new perspective concerning immune metabolic enzymes for understanding the immune system, and offer novel insight into future therapeutic interventions.
Collapse
Affiliation(s)
- Rongrong Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
- School of Life SciencesFudan UniversityShanghaiChina
| | - Xiaobo He
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Jia Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Ganjun Yu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Yanfeng Wu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| |
Collapse
|
2
|
Marzęta-Assas P, Jacenik D, Zasłona Z. Pathophysiology of Arginases in Cancer and Efforts in Their Pharmacological Inhibition. Int J Mol Sci 2024; 25:9782. [PMID: 39337272 PMCID: PMC11431790 DOI: 10.3390/ijms25189782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Arginases are key enzymes that hydrolyze L-arginine to urea and L-ornithine in the urea cycle. The two arginase isoforms, arginase 1 (ARG1) and arginase 2 (ARG2), regulate the proliferation of cancer cells, migration, and apoptosis; affect immunosuppression; and promote the synthesis of polyamines, leading to the development of cancer. Arginases also compete with nitric oxide synthase (NOS) for L-arginine, and their participation has also been confirmed in cardiovascular diseases, stroke, and inflammation. Due to the fact that arginases play a crucial role in the development of various types of diseases, finding an appropriate candidate to inhibit the activity of these enzymes would be beneficial for the therapy of many human diseases. In this review, based on numerous experimental, preclinical, and clinical studies, we provide a comprehensive overview of the biological and physiological functions of ARG1 and ARG2, their molecular mechanisms of action, and affected metabolic pathways. We summarize the recent clinical trials' advances in targeting arginases and describe potential future drugs.
Collapse
Affiliation(s)
| | - Damian Jacenik
- Molecure S.A., 101 Żwirki i Wigury St., 02-089 Warsaw, Poland
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | | |
Collapse
|
3
|
Lee LM, Christodoulou EG, Shyamsunder P, Chen BJ, Lee KL, Fung TK, So CWE, Wong GC, Petretto E, Rackham OJL, Tiong Ong S. A novel network pharmacology approach for leukaemia differentiation therapy using Mogrify ®. Oncogene 2022; 41:5160-5175. [PMID: 36271030 DOI: 10.1038/s41388-022-02505-5] [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: 02/02/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Acute myeloid leukaemia (AML) is a rapidly fatal blood cancer that is characterised by the accumulation of immature myeloid cells in the blood and bone marrow as a result of blocked differentiation. Methods which identify master transcriptional regulators of AML subtype-specific leukaemia cell states and their combinations could be critical for discovering novel differentiation-inducing therapies. In this proof-of-concept study, we demonstrate a novel utility of the Mogrify® algorithm in identifying combinations of transcription factors (TFs) and drugs, which recapitulate granulocytic differentiation of the NB4 acute promyelocytic leukaemia (APL) cell line, using two different approaches. In the first approach, Connectivity Map (CMAP) analysis of these TFs and their target networks outperformed standard approaches, retrieving ATRA as the top hit. We identify dimaprit and mebendazole as a drug combination which induces myeloid differentiation. In the second approach, we show that genetic manipulation of specific Mogrify®-identified TFs (MYC and IRF1) leads to co-operative induction of APL differentiation, as does pharmacological targeting of these TFs using currently available compounds. We also show that loss of IRF1 blunts ATRA-mediated differentiation, and that MYC represses IRF1 expression through recruitment of PML-RARα, the driver fusion oncoprotein in APL, to the IRF1 promoter. Finally, we demonstrate that these drug combinations can also induce differentiation of primary patient-derived APL cells, and highlight the potential of targeting MYC and IRF1 in high-risk APL. Thus, these results suggest that Mogrify® could be used for drug discovery or repositioning in leukaemia differentiation therapy for other subtypes of leukaemia or cancers.
Collapse
MESH Headings
- Humans
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
- Network Pharmacology
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Cell Differentiation/genetics
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Lin Ming Lee
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Eleni G Christodoulou
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Pavithra Shyamsunder
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Bei Jun Chen
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Kian Leong Lee
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Tsz Kan Fung
- Comprehensive Cancer Centre, King's College London, London, UK
- Department of Haematological Medicine, King's College Hospital, London, UK
| | - Chi Wai Eric So
- Comprehensive Cancer Centre, King's College London, London, UK
- Department of Haematological Medicine, King's College Hospital, London, UK
| | - Gee Chuan Wong
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Enrico Petretto
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore.
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
- MRC London Institute of Medical Sciences (LMC), Imperial College London, Faculty of Medicine, London, UK.
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, China.
| | - Owen J L Rackham
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore.
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| | - S Tiong Ong
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
- Department of Haematology, Singapore General Hospital, Singapore, Singapore.
- Department of Medical Oncology, National Cancer Centre, Singapore, Singapore.
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
4
|
OATD-02 Validates the Benefits of Pharmacological Inhibition of Arginase 1 and 2 in Cancer. Cancers (Basel) 2022; 14:cancers14163967. [PMID: 36010962 PMCID: PMC9406419 DOI: 10.3390/cancers14163967] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Arginase 1 and 2 are drivers of multiple immunosuppressive mechanisms and tumour-specific metabolic adaptations. Pharmacological inhibition of extracellular ARG1 has shown antitumour efficacy in various syngeneic tumour models, however, the importance of ARG2 as a therapeutic target has only been demonstrated by genetic deletion studies. This is the first study validating the benefits of pharmacological inhibition of ARG2 in cancer. Our work describes OATD-02 as a potent dual ARG1/ARG2 inhibitor with a cellular activity (necessary for targeting ARG2) exhibiting immunomodulatory and direct antitumour efficacy in animal models. Our results present OATD-02 as an attractive option for combination with other immunotherapeutics, such as PD-1/PD-L1 antibodies or IDO1 inhibitors, especially in the therapy of particularly resistant hypoxic tumours. The presented findings provided the rationale for planning first-in-human clinical trials for OATD-02 in cancer patients. Abstract Background: Arginases play essential roles in metabolic pathways, determining the fitness of both immune and tumour cells. Along with the previously validated role of ARG1 in cancer, the particular significance of ARG2 as a therapeutic target has emerged as its levels correlate with malignant phenotype and poor prognosis. These observations unveil arginases, and specifically ARG2, as well-validated and promising therapeutic targets. OATD-02, a new boronic acid derivative, is the only dual inhibitor, which can address the benefits of pharmacological inhibition of arginase 1 and 2 in cancer. Methods: The inhibitory activity of OATD-02 was determined using recombinant ARG1 and ARG2, as well as in a cellular system using primary hepatocytes and macrophages. In vivo antitumor activity was determined in syngeneic models of colorectal and kidney carcinomas (CT26 and Renca, respectively), as well as in an ARG2-dependent xenograft model of leukaemia (K562). Results: OATD-02 was shown to be a potent dual (ARG1/ARG2) arginase inhibitor with a cellular activity necessary for targeting ARG2. Compared to a reference inhibitor with predominant extracellular activity towards ARG1, we have shown improved and statistically significant antitumor efficacy in the CT26 model and an immunomodulatory effect reflected by Treg inhibition in the Renca model. Importantly, OATD-02 had a superior activity when combined with other immunotherapeutics. Finally, OATD-02 effectively inhibited the proliferation of human K562 leukemic cells both in vitro and in vivo. Conclusions: OATD-02 is a potent small-molecule arginase inhibitor with optimal drug-like properties, including PK/PD profile. Excellent activity against intracellular ARG2 significantly distinguishes OATD-02 from other arginase inhibitors. OATD-02 represents a very promising drug candidate for the combined treatment of tumours, and is the only pharmacological tool that can effectively address the benefits of ARG1/ARG2 inhibition. OATD-02 will enter clinical trials in cancer patients in 2022.
Collapse
|
5
|
Pilanc P, Wojnicki K, Roura AJ, Cyranowski S, Ellert-Miklaszewska A, Ochocka N, Gielniewski B, Grzybowski MM, Błaszczyk R, Stańczak PS, Dobrzański P, Kaminska B. A Novel Oral Arginase 1/2 Inhibitor Enhances the Antitumor Effect of PD-1 Inhibition in Murine Experimental Gliomas by Altering the Immunosuppressive Environment. Front Oncol 2021; 11:703465. [PMID: 34504786 PMCID: PMC8422859 DOI: 10.3389/fonc.2021.703465] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/02/2021] [Indexed: 01/21/2023] Open
Abstract
Glioblastomas (GBM) are the common and aggressive primary brain tumors that are incurable by conventional therapies. Immunotherapy with immune checkpoint inhibitors is not effective in GBM patients due to the highly immunosuppressive tumor microenvironment (TME) restraining the infiltration and activation of cytotoxic T cells. Clinical and experimental studies showed the upregulation of expression of the arginase 1 and 2 (ARG1 and ARG2, respectively) in murine and human GBMs. The elevated arginase activity leads to the depletion of L-arginine, an amino-acid required for the proliferation of T lymphocytes and natural killer cells. Inhibition of ARG1/2 in the TME may unblock T cell proliferation and activate effective antitumor responses. To explore the antitumor potential of ARG1/2 inhibition, we analyzed bulk and single-cell RNA sequencing (scRNA-seq) data from human and murine gliomas. We found the upregulation of ARG1/2 expression in GBMs, both in tumor cells and in tumor infiltrating microglia and monocytes/macrophages. We employed selective arginase inhibitors to evaluate if ARG1/2 inhibition in vitro and in vivo exerts the antitumor effects. A novel, selective ARG1/2 inhibitor - OAT-1746 blocked microglia-dependent invasion of U87-MG and LN18 glioma cells in a Matrigel invasion assay better than reference compounds, without affecting the cell viability. OAT-1746 effectively crossed the blood brain barrier in mice and increased arginine levels in the brains of GL261 glioma bearing mice. We evaluated its antitumor efficacy against GL261 intracranial gliomas as a monotherapy and in combination with the PD-1 inhibition. The oral treatment with OAT-1746 did not affect the immune composition of TME, it induced profound transcriptomic changes in CD11b+ cells immunosorted from tumor-bearing brains as demonstrated by RNA sequencing analyses. Treatment with OAT-1746 modified the TME resulting in reduced glioma growth and increased antitumor effects of the anti-PD-1 antibody. Our findings provide the evidence that inhibition of ARG1/2 activity in tumor cells and myeloid cells in the TME unblocks antitumor responses in myeloid cells and NK cells, and improves the efficacy of the PD-1 inhibition.
Collapse
Affiliation(s)
- Paulina Pilanc
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Salwador Cyranowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Ellert-Miklaszewska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Natalia Ochocka
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartłomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
6
|
Arginase/nitric oxide modifications using live non-pathogenic Leishmania tarentolae as an effective delivery system inside the mammalian macrophages. J Parasit Dis 2021; 45:65-71. [PMID: 33746388 DOI: 10.1007/s12639-020-01279-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022] Open
Abstract
Recombinant live delivery system based on chemokine IFN-γ-inducible protein-10 kDa (CXCL 10 or IP-10), as a suitable immunotherapy tool, have been used for the treatment of Leishmania infections. This chemokine can defeat Leishmania spp. infection via producing nitric oxide (NO) for parasite killing. This study was performed to investigate the effects of IP-10 on the infected human macrophages by L. tarentolae expressing IP-10. We also quantified the arginase activity and NO production in the co-cultured human macrophages with L. tarentolae expressing IP-10 as compared with wild L. tarentolae. The results elucidate that in the infected cells with L. tarentolae expression of IP-10 the arginase activity decreased, and inversely, NO production intensely increased. Altogether, L. tarentolae expressing IP-10 shows a favorable therapeutic tool to improve the treatment of Leishmania infection. This work suggests that L. tarentolae expressing IP-10 cause specific effects on the metabolic pathways of the macrophage host, which might enable the host cells in killing of parasites and decreasing the survival of them against Leishmania infection.
Collapse
|
7
|
S. Clemente G, van Waarde A, F. Antunes I, Dömling A, H. Elsinga P. Arginase as a Potential Biomarker of Disease Progression: A Molecular Imaging Perspective. Int J Mol Sci 2020; 21:E5291. [PMID: 32722521 PMCID: PMC7432485 DOI: 10.3390/ijms21155291] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Arginase is a widely known enzyme of the urea cycle that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The action of arginase goes beyond the boundaries of hepatic ureogenic function, being widespread through most tissues. Two arginase isoforms coexist, the type I (Arg1) predominantly expressed in the liver and the type II (Arg2) expressed throughout extrahepatic tissues. By producing L-ornithine while competing with nitric oxide synthase (NOS) for the same substrate (L-arginine), arginase can influence the endogenous levels of polyamines, proline, and NO•. Several pathophysiological processes may deregulate arginase/NOS balance, disturbing the homeostasis and functionality of the organism. Upregulated arginase expression is associated with several pathological processes that can range from cardiovascular, immune-mediated, and tumorigenic conditions to neurodegenerative disorders. Thus, arginase is a potential biomarker of disease progression and severity and has recently been the subject of research studies regarding the therapeutic efficacy of arginase inhibitors. This review gives a comprehensive overview of the pathophysiological role of arginase and the current state of development of arginase inhibitors, discussing the potential of arginase as a molecular imaging biomarker and stimulating the development of novel specific and high-affinity arginase imaging probes.
Collapse
Affiliation(s)
- Gonçalo S. Clemente
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Inês F. Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands;
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| |
Collapse
|
8
|
Avtandilyan N, Javrushyan H, Mamikonyan A, Grigoryan A, Trchounian A. The potential therapeutic effect of N G-hydroxy-nor-L-arginine in 7,12-dimethylbenz(a)anthracene-induced breast cancer in rats. Exp Mol Pathol 2019; 111:104316. [PMID: 31629728 DOI: 10.1016/j.yexmp.2019.104316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/10/2019] [Accepted: 10/01/2019] [Indexed: 11/19/2022]
Abstract
Advances in our understanding of the metabolism and molecular functions of arginine and their alterations in cancer have led to resurgence in the interest of targeting arginine catabolism as an anticancer strategy. Therefore, arginase inhibitors have been proposed as a way to treat cancer. In this study, the anti-tumor potential of the arginase inhibition by NG-hydroxy-nor-L-arginine (nor-NOHA) (3 mg/kg/day, i.p.), administered for 5 weeks (parallel tumors development, every 3th day) against 7,12-dimethylbenz(a)anthracene (DMBA)-induced mammary carcinogenesis in rats has been investigated. Treatment by nor-NOHA has obvious inhibition effects on development of carcinogenesis in rats was shown. That was seen in downregulation of rats' tumors size and number, mortality rate, in stopped alteration of tissue histopathology, in decrease of polyamines, NO and MDA (malondialdeide) concentrations (in blood). Results have shown arginase and NO-synthase can cooperate to restrain quantities of polyamines and NO for cancer progression. The results obtained can serve as a base to use this model for determination of productive, noncytotoxic antitumor and immune modulating concentration of anticancer agents. Perspectives of targeting arginase and NOS in cancer management can ground application in clinical medicine.
Collapse
Affiliation(s)
- Nikolay Avtandilyan
- Laboratory of Biochemistry, Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia; Department of Biochemistry, Microbiology and Biotechnology, Faculty of Biology, Yerevan State University, Yerevan, Armenia.
| | - Hayarpi Javrushyan
- Laboratory of Biochemistry, Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia.
| | - Anahit Mamikonyan
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Biology, Yerevan State University, Yerevan, Armenia.
| | - Anna Grigoryan
- Department of Zoology, Faculty of Biology, Yerevan State University, Yerevan, Armenia.
| | - Armen Trchounian
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Biology, Yerevan State University, Yerevan, Armenia.
| |
Collapse
|
9
|
Avtandilyan N, Javrushyan H, Karapetyan A, Trchounian A. RETRACTED ARTICLE: Inhibition of Tumor Progression by N G-Nitro-L-arginine Methyl Ester in 7,12- dimethylbenz(a)anthracene Induced Breast Cancer: Nitric Oxide Synthase Inhibition as an Antitumor Prevention. J Mammary Gland Biol Neoplasia 2019; 24:199. [PMID: 30806909 DOI: 10.1007/s10911-019-09428-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 11/28/2022] Open
Affiliation(s)
- Nikolay Avtandilyan
- Laboratory of Biochemistry, Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Republic of Armenia
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Biology, Yerevan State University, 0025, 1 Alex Manoogian, Yerevan, Republic of Armenia
| | - Hayarpi Javrushyan
- Laboratory of Biochemistry, Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Republic of Armenia
| | - Anna Karapetyan
- Department of Human and Animal Physiology, Yerevan State University, Yerevan, Republic of Armenia
| | - Armen Trchounian
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Biology, Yerevan State University, 0025, 1 Alex Manoogian, Yerevan, Republic of Armenia.
| |
Collapse
|