1
|
Bhatia N, Kumar S, Goyal LD, Thareja S. Optimizing selective estrogen receptor degraders for anticancer drug development. Future Med Chem 2025; 17:637-640. [PMID: 40008677 PMCID: PMC11938951 DOI: 10.1080/17568919.2025.2467615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Neha Bhatia
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Lajya Devi Goyal
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, Bathinda, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| |
Collapse
|
2
|
Santovito A, Lambertini M, Nota A. In Vitro and In Vivo Genotoxicity of Polystyrene Microplastics: Evaluation of a Possible Synergistic Action with Bisphenol A. J Xenobiot 2024; 14:1415-1431. [PMID: 39449420 PMCID: PMC11503296 DOI: 10.3390/jox14040079] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
The ubiquitous presence of plastics represents a global threat for all ecosystems and human health. In this study, we evaluated, in vitro and in vivo, the genotoxic potential of different concentrations of polystyrene microplastics (PS-MPs) and their possible synergistic interactions with bisphenol-A (BPA). For the in vitro and the in vivo assays, we used human lymphocytes and hemocytes from Lymnaea stagnalis, respectively. The genomic damage was evaluated by the micronucleus assay, and differences in eggs laid and growth of L. stagnalis were also evaluated. In human lymphocytes, PS-MPs alone at the concentration of 200 μg/mL and in association with BPA 0.100 µg/mL significantly increased the frequencies of micronuclei and nuclear buds, indicating a possible in vitro genotoxic additive action of these two compounds. Vice versa, PS-MPs did not result in genotoxicity in hemocytes. Our results indicated that PS-MPs have genotoxic properties only in vitro and at a concentration of 200 µg/mL; moreover, this compound could intensify the genomic damage when tested with BPA, indicating possible cumulative effects. Finally, PS significantly reduced the growth and the number of laid eggs in L. stagnalis.
Collapse
Affiliation(s)
- Alfredo Santovito
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy
| | - Mattia Lambertini
- Department of Chemistry, University of Turin, Via P. Giuria 7, 10125 Torino, Italy;
| | - Alessandro Nota
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy;
| |
Collapse
|
3
|
Mukai K, Cost R, Zhang XS, Condiff E, Cotton J, Liu X, Boudanova E, Niebel B, Piepenhagen P, Cai X, Park A, Zhou Q. Targeted protein degradation through site-specific antibody conjugation with mannose 6-phosphate glycan. MAbs 2024; 16:2415333. [PMID: 39434219 PMCID: PMC11497922 DOI: 10.1080/19420862.2024.2415333] [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: 04/16/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Recent developments in targeted protein degradation have provided great opportunities to eliminating extracellular protein targets using potential therapies with unique mechanisms of action and pharmacology. Among them, Lysosome-Targeting Chimeras (LYTACs) acting through mannose 6-phosphate receptor (M6PR) have been shown to facilitate degradation of several soluble and membrane-associated proteins in lysosomes with high efficiency. Herein we have developed a novel site-specific antibody conjugation approach to generate antibody mannose 6-phosphate (M6P) conjugates. The method uses a high affinity synthetic M6P glycan, bisM6P, that is coupled to an Fc-engineered antibody NNAS. This mutant without any effector function was generated by switching the native glycosylation site from position 297 to 298 converting non-sialylated structures to highly sialylated N-glycans. The sialic acid of the glycans attached to Asn298 in the engineered antibody was selectively conjugated to bisM6P without chemoenzymatic modification, which is often used for site-specific antibody conjugation through glycans. The conjugate is mainly homogeneous by analysis using mass spectrometry, typically with one or two glycans coupled. The M6P-conjugated antibody against a protein of interest (POI) efficiently internalized targeted soluble proteins, such as human tumor necrosis factor (TNF), in both cancer cell lines and human immune cells, through the endo-lysosomal pathway as demonstrated by confocal microscopy and flow cytometry. TNF in cell culture media was significantly depleted after the cells were incubated with the M6P-conjugated antibody. TNF internalization is mediated through M6PR, and it is correlated well with cell surface expression of cation-independent M6PR (CI-MPR) in immune cells. A significant amount of CI-MPR remains on the cell surface, while internalized TNF is degraded in lysosomes. Thus, the antibody-M6P conjugate is highly efficient in inducing internalization and subsequent lysosome-mediated protein degradation. Our platform provides a unique method for producing biologics-based degraders that may be used to treat diseases through event-driven pharmacology, thereby addressing unmet medical needs.
Collapse
Affiliation(s)
- Kaori Mukai
- Immunology & Inflammation Research, Sanofi, Cambridge, MA, USA
| | - Robert Cost
- Large Molecules Research, Sanofi, Cambridge, MA, USA
| | - Xin Sheen Zhang
- Translational In Vivo Models Research, Sanofi, Cambridge, MA, USA
| | - Emily Condiff
- Translational In Vivo Models Research, Sanofi, Cambridge, MA, USA
| | | | - Xiaohua Liu
- Large Molecules Research, Sanofi, Cambridge, MA, USA
| | | | - Björn Niebel
- Large Molecules Research, Sanofi R&D Ghent, Ghent, Belgium
| | | | - Xinming Cai
- Immunology & Inflammation Research, Sanofi, Cambridge, MA, USA
| | - Anna Park
- Large Molecules Research, Sanofi, Cambridge, MA, USA
| | - Qun Zhou
- Large Molecules Research, Sanofi, Cambridge, MA, USA
| |
Collapse
|
4
|
Kim JY, Park Y, Lee SH, Park E, Lee H. Comparative study on estrogen receptor alpha dimerization and transcriptional activity of parabens. Toxicol Res 2024; 40:153-161. [PMID: 38223674 PMCID: PMC10786792 DOI: 10.1007/s43188-023-00212-1] [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: 02/20/2023] [Revised: 08/27/2023] [Accepted: 09/21/2023] [Indexed: 01/16/2024] Open
Abstract
Parabens are used as preservatives in various household products, including oral products, cosmetics, and hair/body washes. In recent years, the widespread use of parabens has raised concerns due to the potential health risks associated with their estrogenic effects. In the present study, we evaluated and compared the estrogenic activity of parabens using two cell-based in vitro tests: (1) bioluminescence resonance energy transfer (BRET)-based estrogen receptor alpha (ERα) dimerization using HEK293 cells that were stably transfected with ERα-fused NanoLuc luciferase (Nluc) and HaloTag (HT) expression vector, and (2) stably transfected transcriptional activation (STTA) assays using ERα-HeLa9903 cells. The following parabens were tested using the BRET-based ERα dimerization assay and showed estrogenic activity (PC20 values): methyl paraben (MP, 5.98 × 10-5 M), ethyl paraben (EP, 3.29 × 10-5 M), propylparaben (PP, 3.09 × 10-5 M), butyl paraben (BP, 2.58 × 10-5 M), isopropyl paraben (IsoPP, 1.37 × 10-5 M), and isobutyl paraben (IsoBP, 1.43 × 10-5 M). Except MP, all other parabens tested using the STTA assay also showed estrogenic activity: EP, 7.57 × 10-6 M; PP, 1.18 × 10-6 M; BP, 3.02 × 10-7 M; IsoPP, 3.58 × 10-7 M; and IsoBP, 1.80 × 10-7 M. Overall, EP, PP, BP, IsoPP, and IsoBP tested positive for estrogenic activity using both assays. These findings demonstrate that most parabens, albeit not all, induce ERα dimerization and possess estrogenic activity.
Collapse
Affiliation(s)
- Jong-Yeon Kim
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120 Republic of Korea
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326 Republic of Korea
| | - Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang, 10326 Republic of Korea
| | - Eun‐Jung Park
- Department of Food and Nutrition, Gachon University, Seongnam, Gyeonggi‐do 13120 Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, 13120 Republic of Korea
| | - Hae‐Jeung Lee
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120 Republic of Korea
- Department of Food and Nutrition, Gachon University, Seongnam, Gyeonggi‐do 13120 Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, 13120 Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999 Republic of Korea
- Gachon University Gil Medical Center, Incheon, 21565 Republic of Korea
| |
Collapse
|
5
|
Belluti S, Imbriano C, Casarini L. Nuclear Estrogen Receptors in Prostate Cancer: From Genes to Function. Cancers (Basel) 2023; 15:4653. [PMID: 37760622 PMCID: PMC10526871 DOI: 10.3390/cancers15184653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogens are almost ubiquitous steroid hormones that are essential for development, metabolism, and reproduction. They exert both genomic and non-genomic action through two nuclear receptors (ERα and ERβ), which are transcription factors with disregulated functions and/or expression in pathological processes. In the 1990s, the discovery of an additional membrane estrogen G-protein-coupled receptor augmented the complexity of this picture. Increasing evidence elucidating the specific molecular mechanisms of action and opposing effects of ERα and Erβ was reported in the context of prostate cancer treatment, where these issues are increasingly investigated. Although new approaches improved the efficacy of clinical therapies thanks to the development of new molecules targeting specifically estrogen receptors and used in combination with immunotherapy, more efforts are needed to overcome the main drawbacks, and resistance events will be a challenge in the coming years. This review summarizes the state-of-the-art on ERα and ERβ mechanisms of action in prostate cancer and promising future therapies.
Collapse
Affiliation(s)
- Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| |
Collapse
|
6
|
Goel KK, Thapliyal S, Kharb R, Joshi G, Negi A, Kumar B. Imidazoles as Serotonin Receptor Modulators for Treatment of Depression: Structural Insights and Structure-Activity Relationship Studies. Pharmaceutics 2023; 15:2208. [PMID: 37765177 PMCID: PMC10535231 DOI: 10.3390/pharmaceutics15092208] [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: 06/21/2023] [Revised: 08/13/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Serotoninergic signaling is identified as a crucial player in psychiatric disorders (notably depression), presenting it as a significant therapeutic target for treating such conditions. Inhibitors of serotoninergic signaling (especially selective serotonin reuptake inhibitors (SSRI) or serotonin and norepinephrine reuptake inhibitors (SNRI)) are prominently selected as first-line therapy for the treatment of depression, which benefits via increasing low serotonin levels and norepinephrine by blocking serotonin/norepinephrine reuptake and thereby increasing activity. While developing newer heterocyclic scaffolds to target/modulate the serotonergic systems, imidazole-bearing pharmacophores have emerged. The imidazole-derived pharmacophore already demonstrated unique structural characteristics and an electron-rich environment, ultimately resulting in a diverse range of bioactivities. Therefore, the current manuscript discloses such a specific modification and structural activity relationship (SAR) of attempted derivatization in terms of the serotonergic efficacy of the resultant inhibitor. We also featured a landscape of imidazole-based development, focusing on SAR studies against the serotoninergic system to target depression. This study covers the recent advancements in synthetic methodologies for imidazole derivatives and the development of new molecules having antidepressant activity via modulating serotonergic systems, along with their SAR studies. The focus of the study is to provide structural insights into imidazole-based derivatives as serotonergic system modulators for the treatment of depression.
Collapse
Affiliation(s)
- Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University), Haridwar 249404, Uttarakhand, India
| | - Somesh Thapliyal
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
| | - Rajeev Kharb
- Amity Institute of Pharmacy, Amity University, Noida 201313, Uttar Pradesh, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
| | - Arvind Negi
- Department of Bioproduct and Biosystems, Aalto University, 02150 Espoo, Finland
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
- Department of Chemistry, Graphic Era (Deemed to Be University), Dehradun 248002, Uttarakhand, India
| |
Collapse
|
7
|
Minz R, Sharma PK, Negi A, Kesari KK. MicroRNAs-Based Theranostics against Anesthetic-Induced Neurotoxicity. Pharmaceutics 2023; 15:1833. [PMID: 37514018 PMCID: PMC10385075 DOI: 10.3390/pharmaceutics15071833] [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: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Various clinical reports indicate prolonged exposure to general anesthetic-induced neurotoxicity (in vitro and in vivo). Behavior changes (memory and cognition) are compilations commonly cited with general anesthetics. The ability of miRNAs to modulate gene expression, thereby selectively altering cellular functions, remains one of the emerging techniques in the recent decade. Importantly, engineered miRNAs (which are of the two categories, i.e., agomir and antagomir) to an extent found to mitigate neurotoxicity. Utilizing pre-designed synthetic miRNA oligos would be an ideal analeptic approach for intervention based on indicative parameters. This review demonstrates engineered miRNA's potential as prophylactics and/or therapeutics minimizing the general anesthetics-induced neurotoxicity. Furthermore, we share our thoughts regarding the current challenges and feasibility of using miRNAs as therapeutic agents to counteract the adverse neurological effects. Moreover, we discuss the scientific status and updates on the novel neuro-miRNAs related to therapy against neurotoxicity induced by amyloid beta (Aβ) and Parkinson's disease (PD).
Collapse
Affiliation(s)
- Roseleena Minz
- Department of Life Sciences, Central University of Jharkhand, Brambe, Ranchi 853205, Jharkhand, India
| | - Praveen Kumar Sharma
- Department of Life Sciences, Central University of Jharkhand, Brambe, Ranchi 853205, Jharkhand, India
| | - Arvind Negi
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 02150 Espoo, Finland
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, Finland
| |
Collapse
|
8
|
Han X, Sun Y. PROTACs: A novel strategy for cancer drug discovery and development. MedComm (Beijing) 2023; 4:e290. [PMID: 37261210 PMCID: PMC10227178 DOI: 10.1002/mco2.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology has become a powerful strategy in drug discovery, especially for undruggable targets/proteins. A typical PROTAC degrader consists of three components: a small molecule that binds to a target protein, an E3 ligase ligand (consisting of an E3 ligase and its small molecule recruiter), and a chemical linker that hooks first two components together. In the past 20 years, we have witnessed advancement of multiple PROTAC degraders into the clinical trials for anticancer therapies. However, one of the major challenges of PROTAC technology is that only very limited number of E3 ligase recruiters are currently available as E3 ligand for targeted protein degradation (TPD), although human genome encodes more than 600 E3 ligases. Thus, there is an urgent need to identify additional effective E3 ligase recruiters for TPD applications. In this review, we summarized the existing RING-type E3 ubiquitin ligase and their small molecule recruiters that act as effective E3 ligands of PROTAC degraders and their application in anticancer drug discovery. We believe that this review could serve as a reference in future development of efficient E3 ligands of PROTAC technology for cancer drug discovery and development.
Collapse
Affiliation(s)
- Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
- Research Center for Life Science and Human HealthBinjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|
9
|
Site-Specific Antibody Conjugation with Payloads beyond Cytotoxins. Molecules 2023; 28:molecules28030917. [PMID: 36770585 PMCID: PMC9921355 DOI: 10.3390/molecules28030917] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
As antibody-drug conjugates have become a very important modality for cancer therapy, many site-specific conjugation approaches have been developed for generating homogenous molecules. The selective antibody coupling is achieved through antibody engineering by introducing specific amino acid or unnatural amino acid residues, peptides, and glycans. In addition to the use of synthetic cytotoxins, these novel methods have been applied for the conjugation of other payloads, including non-cytotoxic compounds, proteins/peptides, glycans, lipids, and nucleic acids. The non-cytotoxic compounds include polyethylene glycol, antibiotics, protein degraders (PROTAC and LYTAC), immunomodulating agents, enzyme inhibitors and protein ligands. Different small proteins or peptides have been selectively conjugated through unnatural amino acid using click chemistry, engineered C-terminal formylglycine for oxime or click chemistry, or specific ligation or transpeptidation with or without enzymes. Although the antibody protamine peptide fusions have been extensively used for siRNA coupling during early studies, direct conjugations through engineered cysteine or lysine residues have been demonstrated later. These site-specific antibody conjugates containing these payloads other than cytotoxic compounds can be used in proof-of-concept studies and in developing new therapeutics for unmet medical needs.
Collapse
|
10
|
Gupta SM, Behera A, Jain NK, Kumar D, Tripathi A, Tripathi SM, Mujwar S, Patra J, Negi A. Indene-Derived Hydrazides Targeting Acetylcholinesterase Enzyme in Alzheimer's: Design, Synthesis, and Biological Evaluation. Pharmaceutics 2022; 15:pharmaceutics15010094. [PMID: 36678724 PMCID: PMC9860787 DOI: 10.3390/pharmaceutics15010094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
As acetylcholinesterase (AChE) plays a crucial role in advancing Alzheimer's disease (AD), its inhibition is a promising approach for treating AD. Sulindac is an NSAID of the aryl alkanoic acid class, consisting of a indene moiety, which showed neuroprotective behavior in recent studies. In this study, newer Indene analogs were synthesized and evaluated for their in vitro AChE inhibition. Additionally, compared with donepezil as the standard drug, these Indene analogs were accessed for their cell line-based toxicity study on SH-SY5Y cell line. The molecule SD-30, having hydrogen bond donor (HBD) at para-position, showed maximum AChE inhibition potential (IC50 13.86 ± 0.163 µM) in the indene series. Further, the SD-30 showed maximum BuChE inhibition potential (IC50 = 48.55 ± 0.136 µM) with a selectivity ratio of 3.50 and reasonable antioxidant properties compared to ascorbic acid (using DPPH assay). SD-30 (at a dose level: of 10 µM, 20 µM) effectively inhibited AChE-induced Aβ aggregation and showed no significant toxicity up to 30 mM against SH-SY5Y cell lines.
Collapse
Affiliation(s)
- Shraddha Manish Gupta
- Faculty of Pharmacy, Oriental University, Indore 453555, India
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Dehradun 248007, India
| | - Ashok Behera
- Faculty of Pharmacy, DIT University, Dehradun 248009, India
| | - Neetesh K. Jain
- Faculty of Pharmacy, Oriental University, Indore 453555, India
| | - Devendra Kumar
- Faculty of Pharmacy, DIT University, Dehradun 248009, India
| | - Avanish Tripathi
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Shailesh Mani Tripathi
- School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer 305817, India
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India
| | - Jeevan Patra
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Dehradun 248007, India
| | - Arvind Negi
- Faculty of Pharmacy, DIT University, Dehradun 248009, India
- Department of Bioproduct and Biosystems, Aalto University, FI-00076 Espoo, Finland
- Correspondence: or
| |
Collapse
|