1
|
Del Santos N, Vázquez-Ramírez R, Mendes E, Silva Júnior PI, Borges MM. Impact of Mygalin on Inflammatory Response Induced by Toll-like Receptor 2 Agonists and IFN-γ Activation. Int J Mol Sci 2024; 25:10555. [PMID: 39408882 PMCID: PMC11476598 DOI: 10.3390/ijms251910555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Several natural products are being studied to identify new bioactive molecules with therapeutic potential for infections, immune modulation, and other pathologies. TLRs are a family of receptors that play a crucial role in the immune system, constituting the first line of immune defense. They recognize specific products derived from microorganisms that activate multiple pathways and transcription factors in target cells, which are vital for producing immune mediators. Mygalin is a synthetic acylpolyamine derived from hemocytes of the spider Acanthoscurria gomesiana. This molecule negatively regulates macrophage response to LPS stimulation by interacting with MD2 in the TLR4/MD2 complex. Here, we investigated the activity of Mygalin mediated by TLR2 agonists in cells treated with Pam3CSK4 (TLR2/1), Pam2CSK4, Zymosan (TLR2/6), and IFN-γ. Our data showed that Mygalin significantly inhibited stimulation with agonists and IFN-γ, reducing NO and IL-6 synthesis, regardless of the stimulation. There was also a significant reduction in the phosphorylation of proteins NF-κB p65 and STAT-1 in cells treated with Pam3CSK4. Molecular docking assays determined the molecular structure of Mygalin and agonists Pam3CSK4, Pam2CSK4, and Zymosan, as well as their interaction and free energy with the heterodimeric complexes TLR2/1 and TLR2/6. Mygalin interacted with the TLR1 and TLR2 dimer pathway through direct interaction with the agonists, and the ligand-binding domain was similar in both complexes. However, the binding of Mygalin was different from that of the agonists, since the interaction energy with the receptors was lower than with the agonists for their receptors. In conclusion, this study showed the great potential of Mygalin as a potent natural inhibitor of TLR2/1 and TLR2/6 and a suppressor of the inflammatory response induced by TLR2 agonists, in part due to its ability to interact with the heterodimeric complexes.
Collapse
Affiliation(s)
- Nayara Del Santos
- Bacteriology Laboratory, Butantan Institute, São Paulo 05585-000, Brazil;
| | - Ricardo Vázquez-Ramírez
- Institute of Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Elizabeth Mendes
- Bacteriology Laboratory, Butantan Institute, São Paulo 05585-000, Brazil;
| | | | | |
Collapse
|
2
|
Chen YH, Wu KH, Wu HP. Unraveling the Complexities of Toll-like Receptors: From Molecular Mechanisms to Clinical Applications. Int J Mol Sci 2024; 25:5037. [PMID: 38732254 PMCID: PMC11084218 DOI: 10.3390/ijms25095037] [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: 03/28/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
Toll-like receptors (TLRs) are vital components of the innate immune system, serving as the first line of defense against pathogens by recognizing a wide array of molecular patterns. This review summarizes the critical roles of TLRs in immune surveillance and disease pathogenesis, focusing on their structure, signaling pathways, and implications in various disorders. We discuss the molecular intricacies of TLRs, including their ligand specificity, signaling cascades, and the functional consequences of their activation. The involvement of TLRs in infectious diseases, autoimmunity, chronic inflammation, and cancer is explored, highlighting their potential as therapeutic targets. We also examine recent advancements in TLR research, such as the development of specific agonists and antagonists, and their application in immunotherapy and vaccine development. Furthermore, we address the challenges and controversies surrounding TLR research and outline future directions, including the integration of computational modeling and personalized medicine approaches. In conclusion, TLRs represent a promising frontier in medical research, with the potential to significantly impact the development of novel therapeutic strategies for a wide range of diseases.
Collapse
Affiliation(s)
- Yi-Hsin Chen
- Department of Nephrology, Taichung Tzu Chi Hospital, Taichung 427, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Department of Artificial Intelligence and Data Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 613016, Taiwan
| |
Collapse
|
3
|
Shareef U, Altaf A, Ahmed M, Akhtar N, Almuhayawi MS, Al Jaouni SK, Selim S, Abdelgawad MA, Nagshabandi MK. A comprehensive review of discovery and development of drugs discovered from 2020-2022. Saudi Pharm J 2024; 32:101913. [PMID: 38204591 PMCID: PMC10777120 DOI: 10.1016/j.jsps.2023.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/09/2023] [Indexed: 01/12/2024] Open
Abstract
To fully evaluate and define the new drug molecule for its pharmacological characteristics and toxicity profile, pre-clinical and clinical studies are conducted as part of the drug research and development process. The average time required for all drug development processes to finish various regulatory evaluations ranges from 11.4 to 13.5 years, and the expense of drug development is rising quickly. The development in the discovery of newer novel treatments is, however, largely due to the growing need for new medications. Methods to identify Hits and discovery of lead compounds along with pre-clinical studies have advanced, and one example is the introduction of computer-aided drug design (CADD), which has greatly shortened the time needed for the drug to go through the drug discovery phases. The pharmaceutical industry will hopefully be able to address the present and future issues and will continue to produce novel molecular entities (NMEs) to satisfy the expanding unmet medical requirements of the patients as the success rate of the drug development processes is increasing. Several heterocyclic moieties have been developed and tested against many targets and proved to be very effective. In-depth discussion of the drug design approaches of newly found drugs from 2020 to 2022, including their pharmacokinetic and pharmacodynamic profiles and in-vitro and in-vivo assessments, is the main goal of this review. Considering the many stages these drugs are going through in their clinical trials, this investigation is especially pertinent. It should be noted that synthetic strategies are not discussed in this review; instead, they will be in a future publication.
Collapse
Affiliation(s)
- Usman Shareef
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad 44000, Pakistan
| | - Aisha Altaf
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad 44000, Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad 44000, Pakistan
| | - Nosheen Akhtar
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 43600, Pakistan
| | - Mohammed S. Almuhayawi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed K. Nagshabandi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, University of Jeddah, Jeddah 23218, Saudi Arabia
| |
Collapse
|
4
|
Tsukidate T, Hespen CW, Hang HC. Small molecule modulators of immune pattern recognition receptors. RSC Chem Biol 2023; 4:1014-1036. [PMID: 38033733 PMCID: PMC10685800 DOI: 10.1039/d3cb00096f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/03/2023] [Indexed: 12/02/2023] Open
Abstract
Pattern recognition receptors (PRRs) represent a re-emerging class of therapeutic targets for vaccine adjuvants, inflammatory diseases and cancer. In this review article, we summarize exciting developments in discovery and characterization of small molecule PRR modulators, focusing on Toll-like receptors (TLRs), NOD-like receptors (NLRs) and the cGAS-STING pathway. We also highlight PRRs that are currently lacking small molecule modulators and opportunities for chemical biology and therapeutic discovery.
Collapse
Affiliation(s)
- Taku Tsukidate
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
| | - Charles W Hespen
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
| | - Howard C Hang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York New York 10065 USA
- Department of Immunology and Microbiology and Department of Chemistry, Scripps Research, La Jolla California 92037 USA
| |
Collapse
|
5
|
Mills LR, Di Mare F, Gygi D, Lee H, Simmons EM, Kim J, Wisniewski SR, Chirik PJ. Phenoxythiazoline (FTz)-Cobalt(II) Precatalysts Enable C(sp 2 )-C(sp 3 ) Bond-Formation for Key Intermediates in the Synthesis of Toll-like Receptor 7/8 Antagonists. Angew Chem Int Ed Engl 2023:e202313848. [PMID: 37917119 DOI: 10.1002/anie.202313848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/03/2023]
Abstract
Evaluation of the relative rates of the cobalt-catalyzed C(sp2 )-C(sp3 ) Suzuki-Miyaura cross-coupling between the neopentylglycol ester of 4-fluorophenylboronic acid and N-Boc-4-bromopiperidine established that smaller N-alkyl substituents on the phenoxyimine (FI) supporting ligand accelerated the overall rate of the reaction. This trend inspired the design of optimal cobalt catalysts with phenoxyoxazoline (FOx) and phenoxythiazoline (FTz) ligands. An air-stable cobalt(II) precatalyst, (FTz)CoBr(py)3 was synthesized and applied to the cross-coupling of an indole-5-boronic ester nucleophile with a piperidine-4-bromide electrophile that is relevant to the synthesis of reported toll-like receptor (TLR) 7/8 antagonist molecules including afimetoran. Addition of excess KOMe⋅B(Oi Pr)3 improved catalyst lifetime due to attenuation of alkoxide basicity that otherwise resulted in demetallation of the FI chelate. A first-order dependence on the cobalt precatalyst and a saturation regime in nucleophile were observed, supporting turnover-limiting transmetalation and the origin of the observed trends in N-imine substitution.
Collapse
Affiliation(s)
- L Reginald Mills
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Francesca Di Mare
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - David Gygi
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, NJ 08903, USA
| | - Heejun Lee
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, NJ 08903, USA
| | - Eric M Simmons
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, NJ 08903, USA
| | - Junho Kim
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Steven R Wisniewski
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, NJ 08903, USA
| | - Paul J Chirik
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
6
|
Ishizaka ST, Hawkins L, Chen Q, Tago F, Yagi T, Sakaniwa K, Zhang Z, Shimizu T, Shirato M. A novel Toll-like receptor 7/8-specific antagonist E6742 ameliorates clinically relevant disease parameters in murine models of lupus. Eur J Pharmacol 2023; 957:175962. [PMID: 37544422 DOI: 10.1016/j.ejphar.2023.175962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
The sensing of self RNA by the endosomal Toll-like receptors (TLRs) 7 and 8 initiates pathogenic mechanisms underlying the autoimmune disease lupus. A blockade of the TLR7/8 signals may, therefore, be a novel therapeutic intervention for lupus. To test the hypothesis, a novel compound E6742 that blocks TLR7/8 activation was identified. The mode of action of E6742 was investigated by analysis of the tertiary structure of TLR7 and 8 in complex with E6742. The in vitro activities of the compound were examined in cellular systems and its therapeutic potential was evaluated in murine lupus models. Tertiary structures of the extracellular domain of TLR7 and 8 in complex with E6742 showed that E6742 binds specifically and non-covalently to the hydrophobic pocket located at the interface of TLR7 or TLR8 homodimers. E6742 potently and selectively inhibited several TLR7/8-mediated cytokine responses in human PBMC. In two mouse models of lupus, oral dosing of E6742 after the onset of disease suppressed increase in autoantibodies and blocked the advance of organ damage. Collectively, the data show that TLR7/8 activation contributes to disease progression and its blocking by E6742 has potential as a therapeutic intervention for lupus.
Collapse
Affiliation(s)
- Sally T Ishizaka
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | - Lynn Hawkins
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | - Qian Chen
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | | | | | - Kentaro Sakaniwa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Zhikuan Zhang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Manabu Shirato
- Eisai Co., Ltd., Tsukuba Research Laboratories, Ibaraki, Japan.
| |
Collapse
|
7
|
Zheng H, Wu P, Bonnet PA. Recent Advances on Small-Molecule Antagonists Targeting TLR7. Molecules 2023; 28:molecules28020634. [PMID: 36677692 PMCID: PMC9865772 DOI: 10.3390/molecules28020634] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is a class of pattern recognition receptors (PRRs) recognizing the pathogen-associated elements and damage and as such is a major player in the innate immune system. TLR7 triggers the release of pro-inflammatory cytokines or type-I interferons (IFN), which is essential for immunoregulation. Increasing reports also highlight that the abnormal activation of endosomal TLR7 is implicated in various immune-related diseases, carcinogenesis as well as the proliferation of human immunodeficiency virus (HIV). Hence, the design and development of potent and selective TLR7 antagonists based on small molecules or oligonucleotides may offer new tools for the prevention and management of such diseases. In this review, we offer an updated overview of the main structural features and therapeutic potential of small-molecule antagonists of TLR7. Various heterocyclic scaffolds targeting TLR7 binding sites are presented: pyrazoloquinoxaline, quinazoline, purine, imidazopyridine, pyridone, benzanilide, pyrazolopyrimidine/pyridine, benzoxazole, indazole, indole, and quinoline. Additionally, their structure-activity relationships (SAR) studies associated with biological activities and protein binding modes are introduced.
Collapse
Affiliation(s)
- Haoyang Zheng
- Faculty of Pharmacy, Montpellier University, 34093 Montpellier, France
| | - Peiyang Wu
- School of Life Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron IBMM, Ecole Nationale Supérieure de Chimie de Montpellier ENSCM, Montpellier University, Centre National de La Recherche Scientifique CNRS, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
8
|
Alexopoulou L. Nucleic acid-sensing toll-like receptors: Important players in Sjögren’s syndrome. Front Immunol 2022; 13:980400. [PMID: 36389822 PMCID: PMC9659959 DOI: 10.3389/fimmu.2022.980400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Sjögren’s syndrome (SS) is a chronic systemic autoimmune disease that affects the salivary and lacrimal glands, as well as other organ systems like the lungs, kidneys and nervous system. SS can occur alone or in combination with another autoimmune disease, such as systemic lupus erythematosus (SLE) or rheumatoid arthritis. The etiology of SS is unknown but recent studies have revealed the implication of the activation of innate immune receptors, including Toll-like receptors (TLRs), mainly through the detection of endogenous nucleic acids, in the pathogenesis of systemic autoimmune diseases. Studies on SS mouse models suggest that TLRs and especially TLR7 that detects single-stranded RNA of microbial or endogenous origin can drive the development of SS and findings in SS patients corroborate those in mouse models. In this review, we will give an overview of the function and signaling of nucleic acid-sensing TLRs, the interplay of TLR7 with TLR8 and TLR9 in the context of autoimmunity, summarize the evidence for the critical role of TLR7 in the pathogenesis of SS and present a possible connection between SARS-CoV-2 and SS.
Collapse
|
9
|
Das N, Bandopadhyay P, Roy S, Sinha BP, Dastidar UG, Rahaman O, Pal S, Ganguly D, Talukdar A. Development, Optimization, and In Vivo Validation of New Imidazopyridine Chemotypes as Dual TLR7/TLR9 Antagonists through Activity-Directed Sequential Incorporation of Relevant Structural Subunits. J Med Chem 2022; 65:11607-11632. [PMID: 35959635 DOI: 10.1021/acs.jmedchem.2c00386] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Undesirable activation of endosomal toll-like receptors TLR7 and TLR9 present in specific immune cells in response to host-derived ligands is implicated in several autoimmune diseases and other contexts of autoreactive inflammation, making them important therapeutic targets. We report a drug development strategy identifying a new chemotype for incorporating relevant structural subunits into the basic imidazopyridine core deemed necessary for potent TLR7 and TLR9 dual antagonism. We established minimal pharmacophoric features in the core followed by hit-to-lead optimization, guided by in vitro and in vivo biological assays and ADME. A ligand-receptor binding hypothesis was proposed, and selectivity studies against TLR8 were performed. Oral absorption and efficacy of lead candidate 42 were established through favorable in vitro pharmacokinetics and in vivo pharmacodynamic studies, with IC50 values of 0.04 and 0.47 μM against TLR9 and TLR7, respectively. The study establishes imidazopyridine as a viable chemotype to therapeutically target TLR9 and TLR7 in relevant clinical contexts.
Collapse
Affiliation(s)
- Nirmal Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Swarnali Roy
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Bishnu Prasad Sinha
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Uddipta Ghosh Dastidar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Oindrila Rahaman
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Sourav Pal
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
10
|
Cauley AN, Ramirez A, Barhate CL, Donnell AF, Khandelwal P, Sezen-Edmonds M, Sherwood TC, Sloane JL, Cavallaro CL, Simmons EM. Ni/Photoredox-Catalyzed C(sp 2)-C(sp 3) Cross-Coupling of Alkyl Pinacolboronates and (Hetero)Aryl Bromides. Org Lett 2022; 24:5663-5668. [PMID: 35920644 DOI: 10.1021/acs.orglett.2c01942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Utilizing quinoline as a mild, catalytic additive, broadly applicable conditions for the Ni/photoredox-catalyzed C(sp2)-C(sp3) cross-coupling of (hetero)aryl bromides and alkyl pinacolboronate esters were developed, which can be applied to both batch and flow reactions. In addition to primary benzylic nucleophiles, both stabilized and nonstabilized secondary alkyl boronic esters are effective coupling partners. Density functional theory calculations suggest that alkyl radical generation occurs from an alkyl-B(pin)-quinoline complex, which may proceed via an energy transfer process.
Collapse
Affiliation(s)
- Anthony N Cauley
- Chemical Process Development, Bristol Myers Squibb, New Brunswick, New Jersey 08903, United States.,Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Antonio Ramirez
- Chemical Process Development, Bristol Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Chandan L Barhate
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Andrew F Donnell
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Purnima Khandelwal
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Melda Sezen-Edmonds
- Chemical Process Development, Bristol Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Trevor C Sherwood
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Jack L Sloane
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Cullen L Cavallaro
- Small Molecule Drug Discovery, Bristol Myers Squibb, Princeton, New Jersey 08543, United States
| | - Eric M Simmons
- Chemical Process Development, Bristol Myers Squibb, New Brunswick, New Jersey 08903, United States
| |
Collapse
|
11
|
Sreekantha RK, Mussari CP, Dodd DS, Pasunoori L, Hegde S, Posy SL, Critton D, Ruepp S, Subramanian M, Salter-Cid LM, Tagore DM, Sarodaya S, Dudhgaonkar S, Poss MA, Schieven GL, Carter PH, Macor JE, Dyckman AJ. Identification of 2-Pyridinylindole-Based Dual Antagonists of Toll-like Receptors 7 and 8 (TLR7/8). ACS Med Chem Lett 2022; 13:812-818. [PMID: 35586440 PMCID: PMC9109266 DOI: 10.1021/acsmedchemlett.2c00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022] Open
Abstract
The toll-like receptors (TLRs) play key roles in activation of the innate immune system. Aberrant activation of TLR7 and TLR8 pathways can occur in the context of autoimmune disorders due to the elevated presence and recognition of self-RNA as activating ligands. Control of this unintended activation via inhibition of TLR7/8 signaling holds promise for the treatment of diseases such as psoriasis, arthritis, and lupus. Optimization of a 2-pyridinylindole series of compounds led to the identification of potent dual inhibitors of TLR7 and TLR8, which demonstrated good selectivity against TLR9 and other family members. The in vitro characterization and in vivo evaluation in rodent pharmacokinetic/pharmacodynamic and efficacy studies of BMS-905 is detailed, along with structural information obtained through X-ray cocrystallographic studies.
Collapse
Affiliation(s)
| | - Christopher P. Mussari
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dharmpal S. Dodd
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Laxman Pasunoori
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Subramanya Hegde
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Shana L. Posy
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - David Critton
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Stefan Ruepp
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Murali Subramanian
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Luisa M. Salter-Cid
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | | | - Sanket Sarodaya
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | | | - Michael A. Poss
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Gary L. Schieven
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Percy H. Carter
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - John E. Macor
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Alaric J. Dyckman
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
12
|
Betschart C, Faller M, Zink F, Hemmig R, Blank J, Vangrevelinghe E, Bourrel M, Glatthar R, Behnke D, Barker K, Heizmann A, Angst D, Nimsgern P, Jacquier S, Junt T, Zipfel G, Ruzzante G, Loetscher P, Limonta S, Hawtin S, Andre CB, Boulay T, Feifel R, Knoepfel T. Structure-Based Optimization of a Fragment-like TLR8 Binding Screening Hit to an In Vivo Efficacious TLR7/8 Antagonist. ACS Med Chem Lett 2022; 13:658-664. [PMID: 35450354 PMCID: PMC9014506 DOI: 10.1021/acsmedchemlett.1c00696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/08/2022] [Indexed: 11/28/2022] Open
Abstract
Inappropriate activation of TLR7 and TLR8 is linked to several autoimmune diseases, such as lupus erythematosus. Here we report on the efficient structure-based optimization of the inhibition of TLR8, starting from a co-crystal structure of a small screening hit. Further optimization of the physicochemical properties for cellular potency and expansion of the structure-activity relationship for dual potency finally resulted in a highly potent TLR7/8 antagonist with demonstrated in vivo efficacy after oral dosing.
Collapse
Affiliation(s)
- Claudia Betschart
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Michael Faller
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Florence Zink
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - René Hemmig
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jutta Blank
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Eric Vangrevelinghe
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Marjorie Bourrel
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Ralf Glatthar
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Dirk Behnke
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Kerstin Barker
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Andreas Heizmann
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Daniela Angst
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Pierre Nimsgern
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Sébastien Jacquier
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Tobias Junt
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Géraldine Zipfel
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Giulia Ruzzante
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Pius Loetscher
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Sarah Limonta
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Stuart Hawtin
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Cedric Bernard Andre
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Thomas Boulay
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Roland Feifel
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Thomas Knoepfel
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
13
|
Mills LR, Gygi D, Ludwig JR, Simmons EM, Wisniewski SR, Kim J, Chirik PJ. Cobalt-Catalyzed C(sp 2)-C(sp 3) Suzuki-Miyaura Cross-Coupling Enabled by Well-Defined Precatalysts with L,X-Type Ligands. ACS Catal 2022; 12:1905-1918. [PMID: 36034100 PMCID: PMC9400687 DOI: 10.1021/acscatal.1c05586] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cobalt(II) halides in combination with phenoxy-imine (FI) ligands generated efficient precatalysts in situ for the C(sp2)-C(sp3) Suzuki-Miyaura cross coupling between alkyl bromides and neopentylglycol (hetero)arylboronic esters. The protocol enabled efficient C-C bond formation with a host of nucleophiles and electrophiles (36 examples, 34-95%) with precatalyst loadings of 5 mol%. Studies with alkyl halide electrophiles that function as radical clocks support the intermediacy of alkyl radicals during the course of the catalytic reaction. The improved performance of the FI-cobalt catalyst was correlated with decreased lifetimes of cage-escaped radicals as compared to diamine-type ligands. Studies of the phenoxy(imine)-cobalt coordination chemistry validate the L,X interaction leading to the discovery of an optimal, well defined, air-stable mono-FI cobalt(II) precatalyst structure.
Collapse
Affiliation(s)
- L. Reginald Mills
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - David Gygi
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, New Jersey 08903, USA
| | - Jacob R. Ludwig
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Eric M. Simmons
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, New Jersey 08903, USA
| | - Steven R. Wisniewski
- Chemical Process Development, Bristol Myers Squibb Company, New Brunswick, New Jersey 08903, USA
| | - Junho Kim
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Paul J. Chirik
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
14
|
Toll-like Receptor 7 and 8 Imidazoquinoline-Based Agonist/Antagonist Pairs. Bioorg Med Chem Lett 2022; 59:128548. [DOI: 10.1016/j.bmcl.2022.128548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/28/2022]
|
15
|
Dolšak A, Šribar D, Scheffler A, Grabowski M, Švajger U, Gobec S, Holze J, Weindl G, Wolber G, Sova M. Further hit optimization of 6-(trifluoromethyl)pyrimidin-2-amine based TLR8 modulators: Synthesis, biological evaluation and structure-activity relationships. Eur J Med Chem 2021; 225:113809. [PMID: 34488023 DOI: 10.1016/j.ejmech.2021.113809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 8 (TLR8) is an endosomal TLR that has an important role in the innate human immune system, which is involved in numerous pathological conditions. Excessive activation of TLR8 can lead to inflammatory and autoimmune diseases, which highlights the need for development of TLR8 modulators. However, only a few small-molecule modulators that selectively target TLR8 have been developed. Here, we report the synthesis and systematic investigation of the structure-activity relationships of a series of novel TLR8 negative modulators based on previously reported 6-(trifluoromethyl)pyrimidin-2-amine derivatives. Four compounds showed low-micromolar concentration-dependent inhibition of TLR8-mediated signaling in HEK293 cells. These data confirm that the 6-trifluoromethyl group and two other substituents on positions 2 and 4 are important structural elements of pyrimidine-based TLR8 modulators. Substitution of the main scaffold at position 2 with a methylsulfonyl group or para hydroxy/hydroxymethyl substituted benzylamine is essential for potent negative modulation of TLR8. Our best-in-class TLR8-selective modulator 53 with IC50 value of 6.2 μM represents a promising small-molecule chemical probe for further optimization to a lead compound with potent immunomodulatory properties.
Collapse
Affiliation(s)
- Ana Dolšak
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva c. 7, SI-1000, Ljubljana, Slovenia
| | - Dora Šribar
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Alexander Scheffler
- Pharmacology and Toxicology Section, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, 53121, Bonn, Germany
| | - Maria Grabowski
- Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany
| | - Urban Švajger
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000, Ljubljana, Slovenia
| | - Stanislav Gobec
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva c. 7, SI-1000, Ljubljana, Slovenia
| | - Janine Holze
- Pharmacology and Toxicology Section, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, 53121, Bonn, Germany
| | - Günther Weindl
- Pharmacology and Toxicology Section, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, 53121, Bonn, Germany.
| | - Gerhard Wolber
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195, Berlin, Germany.
| | - Matej Sova
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva c. 7, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
16
|
Targeting Toll-like Receptor (TLR) Pathways in Inflammatory Arthritis: Two Better Than One? Biomolecules 2021; 11:biom11091291. [PMID: 34572504 PMCID: PMC8464963 DOI: 10.3390/biom11091291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory arthritis is a cluster of diseases caused by unregulated activity of the immune system. The lost homeostasis is followed by the immune attack of one’s self, what damages healthy cells and tissues and leads to chronic inflammation of various tissues and organs (e.g., joints, lungs, heart, eyes). Different medications to control the excessive immune response are in use, however, drug resistances, flare-reactions and adverse effects to the current therapies are common in the affected patients. Thus, it is essential to broaden the spectrum of alternative treatments and to develop disease-modifying drugs. In the last 20 years, the involvement of the innate immune receptors TLRs in inflammatory arthritis has been widely investigated and targeting either the receptor itself or the proteins in the downstream signalling cascades has emerged as a promising therapeutic strategy. Yet, concerns about the use of pharmacological agents that inhibit TLR activity and may leave the host unprotected against invading pathogens and toxicity issues amid inhibition of downstream kinases crucial in various cellular functions have arisen. This review summarizes the existing knowledge on the role of TLRs in inflammatory arthritis; in addition, the likely druggable related targets and the developed inhibitors, and discusses the pros and cons of their potential clinical use.
Collapse
|
17
|
Beutner GL, Simmons EM, Ayers S, Bemis CY, Goldfogel MJ, Joe CL, Marshall J, Wisniewski SR. A Process Chemistry Benchmark for sp 2-sp 3 Cross Couplings. J Org Chem 2021; 86:10380-10396. [PMID: 34255510 DOI: 10.1021/acs.joc.1c01073] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As sp2-sp3 disconnections gain acceptance in the medicinal chemist's toolbox, an increasing number of potential drug candidates containing this motif are moving into the pharmaceutical development pipeline. This raises a new set of questions and challenges around the novel, direct methodologies available for forging these bonds. These questions gain further importance in the context of process chemistry, where the focus is the development of scalable processes that enable the large-scale delivery of clinical supplies. In this paper, we describe our efforts to apply a wide variety of standard, photo-, and electrochemical sp2-sp3 cross-coupling methods to a pharmaceutically relevant intermediate and optimize each through a combination of high throughput and mechanistically guided experimentation. With data regarding the performance, benefits, and limitations of these novel methods, we evaluate them against a more traditional two-step palladium-catalyzed process. This work reveals trends and similarities between these sp2-sp3 bond-forming methods and suggests a path forward for further refinements.
Collapse
Affiliation(s)
- Gregory L Beutner
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Eric M Simmons
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Sloan Ayers
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Christopher Y Bemis
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Matthew J Goldfogel
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Candice L Joe
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Jonathan Marshall
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Steven R Wisniewski
- Chemical Process Development, Bristol Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| |
Collapse
|
18
|
Kundu B, Raychaudhuri D, Mukherjee A, Sinha BP, Sarkar D, Bandopadhyay P, Pal S, Das N, Dey D, Ramarao K, Nagireddy K, Ganguly D, Talukdar A. Systematic Optimization of Potent and Orally Bioavailable Purine Scaffold as a Dual Inhibitor of Toll-Like Receptors 7 and 9. J Med Chem 2021; 64:9279-9301. [PMID: 34142551 DOI: 10.1021/acs.jmedchem.1c00532] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Several toll-like receptors (TLRs) reside inside endosomes of specific immune cells-among them, aberrant activation of TLR7 and TLR9 is implicated in myriad contexts of autoimmune diseases, making them promising therapeutic targets. However, small-molecule TLR7 and TLR9 antagonists are not yet available for clinical use. We illustrate here the importance of C2, C6, and N9 substitutions in the purine scaffold for antagonism to TLR7 and TLR9 through structure-activity relationship studies using cellular reporter assays and functional studies on primary human immune cells. Further in vitro and in vivo pharmacokinetic studies identified an orally bioavailable lead compound 29, with IC50 values of 0.08 and 2.66 μM against TLR9 and TLR7, respectively. Isothermal titration calorimetry excluded direct TLR ligand-antagonist interactions. In vivo antagonism efficacy against mouse TLR9 and therapeutic efficacy in a preclinical murine model of psoriasis highlighted the potential of compound 29 as a therapeutic candidate in relevant autoimmune contexts.
Collapse
Affiliation(s)
- Biswajit Kundu
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Deblina Raychaudhuri
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India
| | - Ayan Mukherjee
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | | | - Dipika Sarkar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sourav Pal
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Nirmal Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Debdeep Dey
- Tata Medical Center, Newtown, Kolkata 700160, West Bengal, India
| | - Kantubhukta Ramarao
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Kasireddy Nagireddy
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
19
|
Talukdar A, Ganguly D, Roy S, Das N, Sarkar D. Structural Evolution and Translational Potential for Agonists and Antagonists of Endosomal Toll-like Receptors. J Med Chem 2021; 64:8010-8041. [PMID: 34107682 DOI: 10.1021/acs.jmedchem.1c00300] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are members of a large family of evolutionarily conserved pattern recognition receptors (PRRs), which serve as key components of the innate immune system by playing a pivotal role in sensing "nonself" ligands. Endosomal TLRs (TLR3, TLR7, TLR8, and TLR9) can recognize pathogen-derived nucleic acid and initiate an innate immune response because they react against both self- and non-self-origin nucleic acid molecules. Accordingly, both receptor agonists and antagonists are potentially useful in disparate clinical contexts and thus are globally sought after. Recent research has revealed that agonists and antagonists share an overlapping binding region. This Perspective highlights rational medicinal chemistry approaches to elucidate the structural attributes of small molecules capable of agonism or antagonism or of elegantly switching between the two. The structural evolution of different chemotypes can provide the framework for the future development of endosomal TLR agonists and antagonists.
Collapse
Affiliation(s)
- Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata 700091, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Swarnali Roy
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Nirmal Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India.,Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Dipika Sarkar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| |
Collapse
|
20
|
Johnson MB, Chandler M, Afonin KA. Nucleic acid nanoparticles (NANPs) as molecular tools to direct desirable and avoid undesirable immunological effects. Adv Drug Deliv Rev 2021; 173:427-438. [PMID: 33857556 PMCID: PMC8178219 DOI: 10.1016/j.addr.2021.04.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Nucleic acid nanoparticles (NANPs) represent a highly versatile molecular platform for the targeted delivery of various therapeutics. However, despite their promise, further clinical translation of this innovative technology can be hindered by immunological off-target effects. All human cells are equipped with an arsenal of receptors that recognize molecular patterns specific to foreign nucleic acids and understanding the rules that guide this recognition offer the key rationale for the development of therapeutic NANPs with tunable immune stimulation. Numerous recent studies have provided increasing evidence that in addition to NANPs' physicochemical properties and therapeutic effects, their interactions with cells of the immune system can be regulated through multiple independently programmable architectural parameters. The results further suggest that defined immunomodulation by NANPs can either support their immunoquiescent delivery or be used for conditional stimulation of beneficial immunological responses.
Collapse
Affiliation(s)
- M Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Morgan Chandler
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
21
|
Wang X, Liu Y, Han X, Zou G, Zhu W, Shen H, Liu H. Small molecule approaches to treat autoimmune and inflammatory diseases (Part II): Nucleic acid sensing antagonists and inhibitors. Bioorg Med Chem Lett 2021; 44:128101. [PMID: 33984476 DOI: 10.1016/j.bmcl.2021.128101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
Nucleic acid sensing pathways play an important role in the innate immune system, protecting hosts against infections. However, a large body of evidence supports a close association between aberrant activation of those pathways and autoimmune and inflammatory diseases. Part II of the digest series on small molecule approaches to autoimmune and inflammatory diseases concentrates on recent advances with respect to small molecule antagonists or inhibitors of the nucleic acid sensing pathways, including endosomal TLRs, NLRP3 inflammasome and cGAS-STING.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Yafei Liu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xingchun Han
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Ge Zou
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Wei Zhu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Hong Shen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Haixia Liu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China.
| |
Collapse
|
22
|
Prignano F, Lombardo G, Indino S, Ricceri F, Cornaghi L, Donetti EB. Evaluation of expression of Toll-Like Receptors 7 and 9, proliferation, and cytoskeletal biomarkers in plaque and guttate psoriasis: A pilot morphological study. Eur J Histochem 2021; 65. [PMID: 33666385 PMCID: PMC7967266 DOI: 10.4081/ejh.2021.3218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/18/2021] [Indexed: 02/08/2023] Open
Abstract
This pilot study was aimed at comparing TLR7/TLR9 expression, cytoskeletal arrangement, and cell proliferation by indirect immunofluorescence in parallel lesional and non lesional skin samples of guttate psoriasis (PG) and psoriasis vulgaris (PV) in five male patients for each group (n=10). TLR7 expression was detected throughout all the epidermal compartment in PV samples, while in PG skin was restricted to the granular layer. TLR9 was present in the granular layer of non lesional skin and in the suprabasal layers of PV/PG lesional skin. Cell proliferation was localized in all the epidermal layers in lesional PG and PV, consistently with the immunopositivity for the “psoriatic keratin” K16. In the suprabasal layers of lesional PG and PV skin, a similar K17 expression was detected and K10 exhibited a patchy distribution. The present results suggest that TLR7 expression can be considered an intrinsic and differential histomorphological feature of PV.
Collapse
Affiliation(s)
- Francesca Prignano
- Department of Health Sciences, Section of Dermatology, University of Florence.
| | - Giulia Lombardo
- Department of Biomedical Sciences for Health, University of Milan.
| | - Serena Indino
- Department of Biomedical Sciences for Health, University of Milan.
| | - Federica Ricceri
- Department of Health Sciences, Section of Dermatology, University of Florence.
| | - Laura Cornaghi
- Department of Biomedical Sciences for Health, University of Milan.
| | - Elena B Donetti
- Department of Biomedical Sciences for Health, University of Milan.
| |
Collapse
|
23
|
Asami J, Shimizu T. Structural and functional understanding of the toll-like receptors. Protein Sci 2021; 30:761-772. [PMID: 33576548 DOI: 10.1002/pro.4043] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022]
Abstract
Recognition of invading pathogens by the innate immune system is essential to initiate antimicrobial responses and trigger adaptive immunity. This is largely mediated by an array of pattern-recognition receptor families that are essential for recognizing conserved molecular motifs characteristic of pathogenic microbes. One such family is the Toll-like receptors (TLRs). Activation of TLRs induces production of pro-inflammatory cytokines and type I interferons: the former triggers the synthesis of inflammatory mediators which cause fever, pain and other inflammation, and the latter mediates antiviral responses. Over the past decade, significant progress has been made in structural elucidation of TLRs in higher eukaryotes. The TLR structures with and without agonist and antagonist have been revealed by X-ray crystallography and cryo-electron microscopy studies, demonstrating the activated dimer formation induced by the agonistic ligand and the inhibition mechanism of the antagonistic ligand. Intracellular assembled structures and the TLR-chaperone complex are also reported. As the structural understanding of TLRs becomes better integrated with biochemical and immunological studies, a more comprehensive picture of their architectural and functional properties will emerge. This review summarizes recent advances in structural biological and mechanistic studies on TLRs.
Collapse
Affiliation(s)
- Jinta Asami
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Pasquer QTL, Tsakoumagkos IA, Hoogendoorn S. From Phenotypic Hit to Chemical Probe: Chemical Biology Approaches to Elucidate Small Molecule Action in Complex Biological Systems. Molecules 2020; 25:E5702. [PMID: 33287212 PMCID: PMC7730769 DOI: 10.3390/molecules25235702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/22/2023] Open
Abstract
Biologically active small molecules have a central role in drug development, and as chemical probes and tool compounds to perturb and elucidate biological processes. Small molecules can be rationally designed for a given target, or a library of molecules can be screened against a target or phenotype of interest. Especially in the case of phenotypic screening approaches, a major challenge is to translate the compound-induced phenotype into a well-defined cellular target and mode of action of the hit compound. There is no "one size fits all" approach, and recent years have seen an increase in available target deconvolution strategies, rooted in organic chemistry, proteomics, and genetics. This review provides an overview of advances in target identification and mechanism of action studies, describes the strengths and weaknesses of the different approaches, and illustrates the need for chemical biologists to integrate and expand the existing tools to increase the probability of evolving screen hits to robust chemical probes.
Collapse
Affiliation(s)
| | | | - Sascha Hoogendoorn
- Department of Organic Chemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Genève, Switzerland; (Q.T.L.P.); (I.A.T.)
| |
Collapse
|