1
|
Smith TD, Riedl MA. The future of therapeutic options for hereditary angioedema. Ann Allergy Asthma Immunol 2024; 133:380-390. [PMID: 38679158 DOI: 10.1016/j.anai.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
Hereditary angioedema (HAE) is a rare genetic condition causing unpredictable and severe episodes of angioedema that are debilitating and life-threatening. Moreover, HAE can be classified into HAE due to C1-esterase inhibitor deficiency (HAE-C1INH) or HAE with normal C1INH. Moreover, HAE-C1INH is subcategorized as types I and II based on deficient or dysfunctional circulating C1INH protein resulting from inherited or spontaneous mutations in the SERPING1 gene leading to uncontrolled factor XII/plasma kallikrein activation and excessive bradykinin production. Bradykinin-2 receptor activation leads to vasodilation, increased vascular permeability, and smooth muscle contractions, resulting in subcutaneous or submucosal fluid extravasation that can affect the face, extremities, airway, and gastrointestinal and genitourinary systems. Furthermore, HAE with normal C1INH is caused by either a known or unknown genetic mutation, and the mechanisms are less well-established but most forms are thought to be related to bradykinin signaling with a similar presentation as HAE-C1INH despite normal levels of C1INH protein and function. Current HAE management strategies include on-demand and prophylactic treatments which replace C1INH, reduce kallikrein activity, or block bradykinin binding to the bradykinin B2 receptor. With the advent of additional small molecule inhibitors, monoclonal antibodies, RNA-targeted therapies, gene therapies, and gene modification approaches, preclinical studies and human clinical trials are underway to further expand therapeutic options in HAE. This review article will briefly summarize current HAE treatments and provide an overview of potential future therapies for HAE.
Collapse
Affiliation(s)
- Tukisa D Smith
- Division of Allergy, and Immunology, University of California, San Diego, La Jolla, California
| | - Marc A Riedl
- Division of Allergy, and Immunology, University of California, San Diego, La Jolla, California.
| |
Collapse
|
2
|
Stinglhamer M, Kuhlmann JH, Martinelli E, Perulli S, Sandvoss M, Mück-Lichtenfeld C, Derdau V, García Mancheño O. Site-selective Photoredox-Catalyzed Late-stage Benzylic Hydrogen Isotope Exchange. Angew Chem Int Ed Engl 2024:e202411567. [PMID: 39343751 DOI: 10.1002/anie.202411567] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
A highly regioselective visible light photoredox-catalyzed hydrogen isotope exchange (HIE) of benzylic positions in both simple and complex molecules is reported. The process follows a dual catalytic approach using an acridinium photocatalyst in combination with a thiol-based hydrogen atom transfer catalyst, while the use of D2O as an isotope source ensures operational simplicity and cost-effectiveness. High reactivity has been achieved for electron-rich benzylic positions. Moreover, targeted radical formation enables unprecedented selective HIE on intramolecular competing benzylic and alpha to heteroatom positions with moderate to excellent deuterium incorporation. The utility of the reaction was demonstrated on the late-stage HIE of several natural compounds and drug derivatives. Experimental studies and density functional theory (DFT) calculations suggested a single electron transfer (SET) mechanism followed by deprotonation to generate the benzylic radical, and revealed the importance of halogenated solvents or additives. Upon a weak complexation of the halogenated species to the substrate, an oxidation potential lowering effect is induced, as well as a stabilization of the radical-cation species through spin delocalization.
Collapse
Affiliation(s)
- Martin Stinglhamer
- Organic Chemistry Institute, University of Münster, Corrensstraße 36, 48149, Münster, Germany
| | - Jan Hendrik Kuhlmann
- Organic Chemistry Institute, University of Münster, Corrensstraße 36, 48149, Münster, Germany
| | - Elisa Martinelli
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Stefania Perulli
- Organic Chemistry Institute, University of Münster, Corrensstraße 36, 48149, Münster, Germany
| | - Martin Sandvoss
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | | | - Volker Derdau
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Olga García Mancheño
- Organic Chemistry Institute, University of Münster, Corrensstraße 36, 48149, Münster, Germany
| |
Collapse
|
3
|
Do T, Riedl MA. Current and Emerging Therapeutics in Hereditary Angioedema. Immunol Allergy Clin North Am 2024; 44:561-576. [PMID: 38937016 DOI: 10.1016/j.iac.2024.03.009] [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] [Indexed: 06/29/2024]
Abstract
Angioedema is characterized by transient movement of fluid from the vasculature into the interstitial space leading to subcutaneous or submucosal non-pitting edema. Current evidence suggests that most angioedema conditions can be grouped into 2 categories: mast cell-mediated (previously termed histaminergic) or bradykinin-mediated angioedema. Although effective therapies for mast cell-mediated angioedema have existed for decades, specific therapies for bradykinin-mediated angioedema have more recently been developed. In recent years, rigorous studies of these therapies in treating hereditary angioedema (HAE) have led to regulatory approvals of medication for HAE management thereby greatly expanding HAE treatment options.
Collapse
Affiliation(s)
- Toan Do
- Division of Allergy & Immunology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Marc A Riedl
- Division of Allergy & Immunology, University of California, San Diego, 8899 University Center Lane, Suite 230, La Jolla, CA 92122, USA.
| |
Collapse
|
4
|
Riedl MA, Bordone L, Revenko A, Newman KB, Cohn DM. Clinical Progress in Hepatic Targeting for Novel Prophylactic Therapies in Hereditary Angioedema. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:911-918. [PMID: 38142864 DOI: 10.1016/j.jaip.2023.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 12/26/2023]
Abstract
Hereditary angioedema (HAE) is typically caused by a deficiency of the protease inhibitor C1 inhibitor (C1INH). The absence of C1INH activity on plasma kallikrein and factor XIIa leads to overproduction of the vasoactive peptide bradykinin, with resulting angioedema. As the primary site of C1INH and prekallikrein production, the liver is recognized as an important therapeutic target in HAE, leading to the development of hepatic-focused treatment strategies such as GalNAc-conjugated antisense technology and gene modification. This report reviews currently available data on hepatic-focused interventions for HAE that have advanced into human trials. Donidalorsen is an investigational GalNAc3-conjugated antisense oligonucleotide that binds to prekallikrein mRNA in the liver and reduces the expression of prekallikrein. Phase 2 data with subcutaneous donidalorsen demonstrated a significant reduction in HAE attack rate compared with placebo. Phase 3 trials are underway. ADX-324 is a GalNAc3-conjugated short-interfering RNA being investigated in HAE. BMN 331 is an investigational AAV5-based gene therapy vector that expresses wild-type human C1INH and is targeted to hepatocytes. A single intravenous dose of BMN 331 is intended to replace the defective SERPING1 gene and enable patients to produce functional C1INH. A first-in-human phase 1/2 study is ongoing with BMN 331. NTLA-2002 is an investigational in vivo clustered regularly interspaced short palindromic repeats/Cas9-based therapy designed to knock out the prekallikrein-coding KLKB1 gene in hepatocytes; a phase 1/2 study is ongoing. Findings from these and other ongoing studies are highly anticipated with the expectation of expanding the array of treatment options in HAE.
Collapse
Affiliation(s)
- Marc A Riedl
- Division of Allergy and Immunology, University of California, San Diego, La Jolla, Calif.
| | | | | | | | - Danny M Cohn
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Li P, Du Z, Wu B, Zhao X, You Y. Highly effective and selective FeBr 3-promoted deuterium bromination/cyclization of 1, n-enynes. Org Biomol Chem 2024; 22:959-964. [PMID: 38205648 DOI: 10.1039/d3ob01778h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
A highly effective and selective FeBr3-promoted deuterium bromination/cyclization of 1,n-enynes is reported. On the one hand, the Lewis acid FeBr3 as a catalyst promotes cyclization of 1,n-enynes to afford deuterium heterocyclic frameworks with high efficiency. On the other hand, FeBr3 serves as the bromine source (with D2O as the deuterium source) to promote the formation of the desired deuterated pyrrole derivatives containing alkenyl bromide groups. This protocol provides an effective pathway to afford deuterated alkenyl brominative compounds as (Z)-isomers with high yields and selectivity, offering a new method for introducing 2H into organic compounds.
Collapse
Affiliation(s)
- Ping Li
- Department of Cable Engineering, Henan Institute of Technology, Xinxiang, 453000, China
| | - Zhongjian Du
- School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Hefei 230009, China.
| | - Baofeng Wu
- Research Institute of Exploration and Development, PetroChina, Daqing Oilfield Company, Daqing 163712, China
| | - Xin Zhao
- Research Institute of Exploration and Development, PetroChina, Daqing Oilfield Company, Daqing 163712, China
| | - Yang'en You
- School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Hefei 230009, China.
| |
Collapse
|
6
|
Di Martino RMC, Maxwell BD, Pirali T. Deuterium in drug discovery: progress, opportunities and challenges. Nat Rev Drug Discov 2023; 22:562-584. [PMID: 37277503 PMCID: PMC10241557 DOI: 10.1038/s41573-023-00703-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 06/07/2023]
Abstract
Substitution of a hydrogen atom with its heavy isotope deuterium entails the addition of one neutron to a molecule. Despite being a subtle change, this structural modification, known as deuteration, may improve the pharmacokinetic and/or toxicity profile of drugs, potentially translating into improvements in efficacy and safety compared with the non-deuterated counterparts. Initially, efforts to exploit this potential primarily led to the development of deuterated analogues of marketed drugs through a 'deuterium switch' approach, such as deutetrabenazine, which became the first deuterated drug to receive FDA approval in 2017. In the past few years, the focus has shifted to applying deuteration in novel drug discovery, and the FDA approved the pioneering de novo deuterated drug deucravacitinib in 2022. In this Review, we highlight key milestones in the field of deuteration in drug discovery and development, emphasizing recent and instructive medicinal chemistry programmes and discussing the opportunities and hurdles for drug developers, as well as the questions that remain to be addressed.
Collapse
Affiliation(s)
| | | | - Tracey Pirali
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
7
|
Vachlioti E, Ferikoglou S, Georgiou X, Karampatsis V, Afratis K, Bafiti V, Savard M, Papaioannou D, Katsila T, Gobeil F, Rassias G. Development of a multigram synthesis of the bradykinin receptor 2 agonist FR-190997 and analogs thereof. Arch Pharm (Weinheim) 2023; 356:e2200610. [PMID: 36720040 DOI: 10.1002/ardp.202200610] [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: 11/28/2022] [Revised: 12/24/2022] [Accepted: 01/13/2023] [Indexed: 02/02/2023]
Abstract
Using Fujisawa's B2R agonist FR-190997, we recently demonstrated for the first time that agonism at the bradykinin receptor type 2 (B2R) produces substantial antiproliferative effects. FR-190997 elicited an EC50 of 80 nM in the triple-negative breast cancer cell line MDA-MB-231, a much superior performance to that exhibited by most approved breast cancer drugs. Consequently, we initiated a program aiming primarily at synthesizing adequate quantities of FR-190997 to support further in vitro and in vivo studies toward its repurposing for various cancers and, in parallel, enable the generation of novel FR-190997 analogs for an SAR study. Prerequisite for this endeavor was to address the synthetic challenges associated with the FR-190997 scaffold, which the Fujisawa chemists had constructed in 20 steps, 13 of which required chromatographic purification. We succeeded in developing a 17-step synthesis amenable to late-stage diversification that eliminated all chromatography and enabled access to multigram quantities of FR-190997 and novel derivatives thereof, supporting further anticancer research based on B2R agonists.
Collapse
Affiliation(s)
- Eleanna Vachlioti
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| | - Spyridon Ferikoglou
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| | - Xenios Georgiou
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| | - Vasilios Karampatsis
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| | - Konstantinos Afratis
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| | - Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Martin Savard
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Fernand Gobeil
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gerasimos Rassias
- Department of Chemistry, University of Patras, Rio University Campus, Patra, Greece
| |
Collapse
|
8
|
Bachelard H, Marceau F. Are noscapine and raloxifene ligands of the bradykinin B 2 receptor? An assessment based on the human umbilical vein contractility assay. Int Immunopharmacol 2022; 110:108984. [PMID: 35780642 DOI: 10.1016/j.intimp.2022.108984] [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: 05/10/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/29/2022]
Abstract
The centrally acting antitussive opiate derivative, noscapine, has been claimed to be a non-competitive bradykinin B2 receptor antagonist. Raloxifene, a selective estrogen receptor modulator, was predicted to bind the bradykinin B2 receptor and to exert a partial agonist activity. These intriguing claims suggest that new molecular scaffolds ("chemotypes") may be identified for small molecule ligands of kinin receptors and that some off-target effects of noscapine or raloxifene may be mediated by bradykinin B2 receptors. An established contractile bioassay for ligands of the bradykinin B2 receptor, the isolated human umbilical vein, was exploited to characterize the inhibitory effect of noscapine and raloxifene on the B2 receptor-mediated contractile response to bradykinin. Observed effects were compared with those of the peptide antagonist icatibant, a potent, selective and competitive B2 receptor antagonist. Our results indicate that neither noscapine (2.5 µM) nor raloxifene (20 µM) behave as B2 receptor antagonists in concentrations that vastly exceeded an effective concentration of the control antagonist, icatibant; further, none of these drugs had direct contractile effects. It is suggested that the previously reported B2 receptor inhibitory effect of noscapine, a putative sigma-receptor agonist, might result from an indirect physiological antagonism, while raloxifene did not appear to have any significant affinity for the B2 receptors.
Collapse
Affiliation(s)
- Hélène Bachelard
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC G1V 4G2, Canada.
| | - François Marceau
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
9
|
Rex DAB, Vaid N, Deepak K, Dagamajalu S, Prasad TSK. A comprehensive review on current understanding of bradykinin in COVID-19 and inflammatory diseases. Mol Biol Rep 2022; 49:9915-9927. [PMID: 35596055 PMCID: PMC9122735 DOI: 10.1007/s11033-022-07539-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/28/2022] [Indexed: 12/28/2022]
Abstract
Bradykinin, a member of the kallikrein–kinin system (KKS), is a potent, short-lived vasoactive peptide that acts as a vasodilator and an inflammatory mediator in a number of signaling mechanisms. Bradykinin induced signaling is mediated through kinin B1 (BDKRB1) and B2 (BDKRB2) transmembrane receptors coupled with different subunits of G proteins (Gαi/Gα0, Gαq and Gβ1γ2). The bradykinin-mediated signaling mechanism activates excessive pro-inflammatory cytokines, including IL-6, IL-1β, IL-8 and IL-2. Upregulation of these cytokines has implications in a wide range of clinical conditions such as inflammation leading to fibrosis, cardiovascular diseases, and most recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In SARS-CoV-2 infection, bradykinin is found to be at raised levels and is reported to trigger a diverse array of symptoms. All of this brings bradykinin to the core point as a molecule of immense therapeutic value. Our understanding of its involvement in various pathways has expanded with time. Therefore, there is a need to look at the overall picture that emerges from the developments made by deciphering the bradykinin mediated signaling mechanisms involved in the pathological conditions. It will help devise strategies for developing better treatment modalities in the implicated diseases. This review summarizes the current state of knowledge on bradykinin mediated signaling in the diverse conditions described above, with a marked emphasis on the therapeutic potential of targeting the bradykinin receptor.
Collapse
Affiliation(s)
- Devasahayam Arokiar Balaya Rex
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Neelanchal Vaid
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - K Deepak
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|