10 research outputs found

    Discovery of novel Fe(II)/α-ketoglutarate-dependent dioxygenases for oxidation of L-proline

    No full text
    Genome-mining for novel Fe(II)/α-ketoglutarate-dependent dioxygenases (αKGDs) to expand the enzymatic repertoire in the oxidation of L-proline is reported. Through clustering of genes, we predicted regio- and stereoselectivity in hydroxylation reaction and validated this hypothesis experimentally. Two novel by-products in reactions with BcePH and Ssp5PH were observed, isolated and structure was determined as an epoxide and a 3,4-diol, respectively. Mechanism for formation of epoxide is suggested and validated by using 18O-labelling experiment, that proceeds via cis-3-hydroxylation step first, followed by ring closure. A Biocatalytic step was performed on sub-gram quantities of starting material without any significant condition optimization. The substrate concentration, however, is already up to 40-fold higher than the usually reported titers for P450-mediated hydroxylations, showing the synthetic potential of αKGDs on preparative scal

    Discovery of small-molecule reversible Factor D inhibitors targeting the alternative complement pathway

    Get PDF
    Complement is a key component of the innate immune system, recognizing pathogens and promoting their elimination through cell lysis and/or inflammatory responses1,2. Activation of complement can be initiated by three distinct routes, the classical, lectin and alternative pathways. These converge at the proteolytic cleavage of the third component of complement, C3, generating the C3a and C3b activation fragments. C3b also participates in amplification of its own production via the positive feedback loop of the alternative pathway (AP)2,3. The two proteases Factor D and Factor B are essential for this tightly regulated amplification process. Dysregulation of AP activity predisposes individuals to diverse disorders including age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS) and C3 nephropathy4-7. Here, using a structure-based design approach, we describe the identification of potent and selective small-molecule reversible inhibitors of Factor D. These inhibitors efficiently blocked AP activation in human whole blood, and prevented both C3 deposition onto, and lysis of, human erythrocytes in an assay that mimics erythrocyte lytic sensitivity. These findings were confirmed with erythrocytes from PNH patients. Oral administration inhibited lipopolysaccharide (LPS)-induced AP activation both systemically and in ocular tissues in Factor D-humanized C57Bl/6 mice. These data demonstrate the feasibility of inhibiting the AP with specific small molecule antagonists and support the development of oral Factor D inhibitors that would enable the systemic treatment of many complement-mediated diseases for which there are currently either no or sub-optimal therapies

    JDQ443, a Structurally Novel, Pyrazole-Based, Covalent Inhibitor of KRASG12C for the Treatment of Solid Tumors.

    No full text
    Rapid emergence of tumor resistance via RAS pathway reactivation has been reported from clinical studies of covalent KRASG12C inhibitors. Thus, inhibitors with broad potential for combination treatment and distinct binding modes to overcome resistance mutations may prove beneficial. JDQ443 is an investigational covalent KRASG12C inhibitor derived from structure-based drug design followed by extensive optimization of two dissimilar prototypes. JDQ443 is a stable atropisomer containing a unique 5-methylpyrazole core and a spiro-azetidine linker designed to position the electrophilic acrylamide for optimal engagement with KRASG12C C12. A substituted indazole at pyrazole position 3 results in novel interactions with the binding pocket that do not involve residue H95. JDQ443 showed PK/PD activity in vivo and dose-dependent antitumor activity in mouse xenograft models. JDQ443 is now in clinical development, with encouraging early phase data reported from an ongoing Phase Ib/II clinical trial (NCT04699188)

    Discovery of Highly Potent and Selective Small-Molecule Reversible Factor D Inhibitors Demonstrating Alternative Complement Pathway Inhibition <i>in Vivo</i>

    No full text
    The highly specific S1 serine protease factor D (FD) plays a central role in the amplification of the complement alternative pathway (AP) of the innate immune system. Genetic associations in humans have implicated AP activation in age-related macular degeneration (AMD), and AP dysfunction predisposes individuals to disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). The combination of structure-based hit identification and subsequent optimization of the center (<i>S</i>)-proline-based lead <b>7</b> has led to the discovery of noncovalent reversible and selective human factor D (FD) inhibitors with drug-like properties. The orally bioavailable compound <b>2</b> exerted excellent potency in 50% human whole blood <i>in vitro</i> and blocked AP activity <i>ex vivo</i> after oral administration to monkeys as demonstrated by inhibition of membrane attack complex (MAC) formation. Inhibitor <b>2</b> demonstrated sustained oral and ocular efficacy in a model of lipopolysaccharide (LPS)-induced systemic AP activation in mice expressing human FD

    Discovery of Highly Potent and Selective Small-Molecule Reversible Factor D Inhibitors Demonstrating Alternative Complement Pathway Inhibition <i>in Vivo</i>

    No full text
    The highly specific S1 serine protease factor D (FD) plays a central role in the amplification of the complement alternative pathway (AP) of the innate immune system. Genetic associations in humans have implicated AP activation in age-related macular degeneration (AMD), and AP dysfunction predisposes individuals to disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). The combination of structure-based hit identification and subsequent optimization of the center (<i>S</i>)-proline-based lead <b>7</b> has led to the discovery of noncovalent reversible and selective human factor D (FD) inhibitors with drug-like properties. The orally bioavailable compound <b>2</b> exerted excellent potency in 50% human whole blood <i>in vitro</i> and blocked AP activity <i>ex vivo</i> after oral administration to monkeys as demonstrated by inhibition of membrane attack complex (MAC) formation. Inhibitor <b>2</b> demonstrated sustained oral and ocular efficacy in a model of lipopolysaccharide (LPS)-induced systemic AP activation in mice expressing human FD

    JDQ443, a Structurally Novel, Pyrazole-Based, Covalent Inhibitor of KRAS<sup>G12C</sup> for the Treatment of Solid Tumors

    No full text
    Rapid emergence of tumor resistance via RAS pathway reactivation has been reported from clinical studies of covalent KRASG12C inhibitors. Thus, inhibitors with broad potential for combination treatment and distinct binding modes to overcome resistance mutations may prove beneficial. JDQ443 is an investigational covalent KRASG12C inhibitor derived from structure-based drug design followed by extensive optimization of two dissimilar prototypes. JDQ443 is a stable atropisomer containing a unique 5-methylpyrazole core and a spiro-azetidine linker designed to position the electrophilic acrylamide for optimal engagement with KRASG12C C12. A substituted indazole at pyrazole position 3 results in novel interactions with the binding pocket that do not involve residue H95. JDQ443 showed PK/PD activity in vivo and dose-dependent antitumor activity in mouse xenograft models. JDQ443 is now in clinical development, with encouraging early phase data reported from an ongoing Phase Ib/II clinical trial (NCT04699188)

    JDQ443, a Structurally Novel, Pyrazole-Based, Covalent Inhibitor of KRAS<sup>G12C</sup> for the Treatment of Solid Tumors

    No full text
    Rapid emergence of tumor resistance via RAS pathway reactivation has been reported from clinical studies of covalent KRASG12C inhibitors. Thus, inhibitors with broad potential for combination treatment and distinct binding modes to overcome resistance mutations may prove beneficial. JDQ443 is an investigational covalent KRASG12C inhibitor derived from structure-based drug design followed by extensive optimization of two dissimilar prototypes. JDQ443 is a stable atropisomer containing a unique 5-methylpyrazole core and a spiro-azetidine linker designed to position the electrophilic acrylamide for optimal engagement with KRASG12C C12. A substituted indazole at pyrazole position 3 results in novel interactions with the binding pocket that do not involve residue H95. JDQ443 showed PK/PD activity in vivo and dose-dependent antitumor activity in mouse xenograft models. JDQ443 is now in clinical development, with encouraging early phase data reported from an ongoing Phase Ib/II clinical trial (NCT04699188)

    Structure-based design and pre-clinical characterization of selective and orally bioavailable Factor XIa inhibitors: Demonstrating the power of an integrated S1 protease family approach

    No full text
    The serine protease Factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anti-coagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement Factor D inhibitor and exhibited sub-micromolar FXIa activity and an encouraging ADME profile while being devoid of peptidomimetic architecture. Optimization of interactions in the S1, S1β, and S1` pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with sub-nanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a pre-clinical PK profile consistent with bid dosing in patients

    Structure-Based Design and Preclinical Characterization of Selective and Orally Bioavailable Factor XIa Inhibitors: Demonstrating the Power of an Integrated S1 Protease Family Approach

    No full text
    The serine protease Factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anti-coagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement Factor D inhibitor and exhibited sub-micromolar FXIa activity and an encouraging ADME profile while being devoid of peptidomimetic architecture. Optimization of interactions in the S1, S1β, and S1` pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with sub-nanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a pre-clinical PK profile consistent with bid dosing in patients

    Small-molecule factor D inhibitors targeting the alternative complement pathway

    No full text
    Complement is a key component of the innate immune system, recognizing pathogens and promoting their elimination through cell lysis and/or inflammatory responses1,2. Activation of complement can be initiated by three distinct routes, the classical, lectin and alternative pathways. These converge at the proteolytic cleavage of the third component of complement, C3, generating the C3a and C3b activation fragments. C3b also participates in amplification of its own production via the positive feedback loop of the alternative pathway (AP)2,3. The two proteases Factor D and Factor B are essential for this tightly regulated amplification process. Dysregulation of AP activity predisposes individuals to diverse disorders including age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS) and C3 nephropathy4-7. Here, using a structure-based design approach, we describe the identification of potent and selective small-molecule reversible inhibitors of Factor D. These inhibitors efficiently blocked AP activation in human whole blood, and prevented both C3 deposition onto, and lysis of, human erythrocytes in an assay that mimics erythrocyte lytic sensitivity. These findings were confirmed with erythrocytes from PNH patients. Oral administration inhibited lipopolysaccharide (LPS)-induced AP activation both systemically and in ocular tissues in Factor D-humanized C57Bl/6 mice. These data demonstrate the feasibility of inhibiting the AP with specific small molecule antagonists and support the development of oral Factor D inhibitors that would enable the systemic treatment of many complement-mediated diseases for which there are currently either no or sub-optimal therapies
    corecore