key: cord-1006313-t8b9ekkg authors: Vandyck, Koen; Deval, Jerome title: Considerations for the Discovery and Development of 3-Chymotrypsin-Like Cysteine Protease Inhibitors Targeting SARS-CoV-2 Infection date: 2021-04-27 journal: Curr Opin Virol DOI: 10.1016/j.coviro.2021.04.006 sha: 86cc3190b0aaf484d5590d5bd7250b108434b423 doc_id: 1006313 cord_uid: t8b9ekkg Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the COVID-19 pandemic. The coronavirus 3-chymotrypsin-like protease (3CLpro) controls virus replication and is therefore considered a major target and promising opportunity for rational-based antiviral discovery with direct acting agents. Here we review first-generation SARS-CoV-2 3CLpro inhibitors PF-07304814, GC-376, and CDI-45205 that are being delivered either by injection or inhalation due to their low intrinsic oral bioavailability. In addition, PF-07321332 is now emerging as a promising second-generation clinical candidate for oral delivery. A key challenge to the development of novel 3CLpro inhibitors is the poor understanding of the predictive value of in vitro potency toward clinical efficacy, an issue complicated by the involvement of host proteases in virus entry. Further preclinical and clinical validation will be key to establishing 3CLpro inhibitors as a bona fide class for future SARS-CoV-2 therapeutics for both hospitalized and outpatient populations. COVID-19 is a respiratory infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although most infected individuals are either asymptomatic or experience selflimiting symptoms similar to the common cold, clinical cases of severe COVID-19 require hospitalization and intensive care due to pneumonia and extra-pulmonary manifestations (Harapan, Itoh et al. 2020 , Wiersinga, Rhodes et al. 2020 . The anti-inflammatory corticosteroid dexamethasone, and nucleotide analog remdesivir are considered the standards of care for the treatment of severe COVID-19 in hospitalized patients needing supplemental oxygen (https://www.covid19treatmentguidelines.nih.gov/therapeutic-management/). However, these molecules are not suitable for patients in the early stage of SARS-CoV-2 infection, in outpatient settings, or as prophylaxis. SARS-CoV-2 is an enveloped, positive-sense, single-stranded RNA virus that belongs to the β-coronavirus genus of the Coronaviridae family (Coronaviridae Study Group of the International Committee on Taxonomy of 2020). The main viral protease, 3CLpro, is a cysteine protease with distinctive substrate preference for glutamine at the P1 site (Leu-Gln/(Ser,Ala,Gly)) (Fan, Ma et al. 2005) . Due to its key role in viral replication, 3CLpro is considered a major target for antiviral drug discovery. Intense research and development are currently ongoing to evaluate 3CLpro inhibitors as potential treatments for COVID-19. The urgent need for COVID-19 treatments triggered rapid repurposing of protease inhibitors previously developed for other viral indications. FDA-approved HIV protease inhibitors do not provide any benefit for COVID-19 patients (Cao, Wang et al. 2020) . The most interesting subgroup of protease inhibitors are peptidomimetics previously designed to inhibit 3C-or 3CL-like proteases. These molecules contain a P1-glutamine surrogate, like those designed for the discovery of the rhinovirus 3Cpro inhibitor rupintrivir (Patick, Brothers et al. 2005) . While rupintrivir itself did not display significant activity against SARS-CoV-2 3CLpro (Liu 2021), peptidomimetics previously described for other coronaviruses were expected to have broad activity across most coronaviruses as their substrate cleavage sites are highly conserved (for review: (Ullrich and Nitsche 2020)). Unsurprisingly, some peptidomimetic compounds active against MERS and SARS-CoV-1 are also active against SARS-CoV-2. PF-00835231 was J o u r n a l P r e -p r o o f developed more than 15 years ago against SARS-CoV-1 (patent WO2005113580) and recently showed high in vitro potency against SARS-CoV-2 (Figure 1 ) (Zhang, Lin et al. 2020 , Boras, Jones et al. 2021 . Currently its phosphate prodrug PF-07304814 is being evaluated in a Phase 1b clinical trial in hospitalized patients with COVID-19; safety and efficacy results are expected around mid-2021 (ClinTrials.gov Identifier NCT04535167). The peptidomimetic compound GC-376 is a broad-spectrum inhibitor of 3C and 3C-like proteases of picornaviruses, noroviruses and coronaviruses (Figure 1 ) (Kim, Lovell et al. 2012) . The compound has been licensed to Anivive, a company focusing on Companion animal veterinary medicine, to treat feline infectious peritonitis caused by a feline coronavirus (Kim, Lovell et al. 2012 , Takahashi, Kim et al. 2013 , Kim, Liu et al. 2016 , Pedersen, Kim et al. 2018 . GC-376 was confirmed to also block SARS-CoV-2 (Gurard-Levin, , Ma, Sacco et al. 2020 , providing a scientific rationale for a pre-IND submission for the treatment COVID-19 (https://www.prnewswire.com/newsreleases/anivive-repurposes-veterinary-drug-gc376-for-covid-19-and-submits-pre-ind-to-fda-301065619.html). GC-376 is a sodium bisulfite prodrug of the aldehyde GC-373. Bisulfite adducts increase chemical stability and/or aqueous solubility of aldehydes. GC-373 has been described as a potent cathepsin B inhibitor (Swisher, Prior et al. 2015) , and its prodrug GC-376 has been described as a potent cathepsin L inhibitor . The low target selectivity of GC-376 and many other 3CLpro inhibitors could present a challenge for clinical development. In particular, 3CLpro inhibitors that lack selectivity toward cathepsin L may potentially have counter-effective side effects by dampening the immune response against SARS-CoV-2 (Baranov, Bianchi et al. 2020) . The broad spectrum of virus protease inhibition previously reported for GC-376 combined with the cathepsin L inhibition effect suggest its activity may extend to other human proteases. CDI-45205 (undisclosed structure) is the latest compound to advance into late preclinical stage (https://www.cocrystalpharma.com/news/pressreleases/detail/105/cocrystal-pharma-selects-lead-compound-for-further). It was selected by Cocrystal Pharma at the end of 2020 from the broad-spectrum protease inhibitors discovered by Kansas State University Research Foundation, which demonstrated in vitro and in vivo activity in animal models against MERS and in vitro activity against SARS-CoV-2 (Rathnayake, Zheng et al. 2020) . Although SARS-CoV-2 infects mainly the respiratory tract (pharynx, trachea, lungs) at the early stages of the self-limiting infection, a broader organotropism has been reported in more severe and advanced cases of illness (Kruger, Gross et al. 2020 , Trypsteen, Van Cleemput et al. 2020 , Zhang, Geng et al. 2020 . In addition to the principal target organs of virus replication, several other factors need to be considered when optimizing the route of administration of a 3CLpro inhibitor, including the timing of intervention relative to SARS-CoV-2 infection, COVID-19 disease stage, and bioavailability of the antiviral agent (Figure 2) . The nucleotide analog remdesivir is approved in the United States for the intravenous infusion treatment of hospitalized patients only; therefore, the need for easier administration of a direct acting agent in early-stage non-hospitalized patients remains high, as is the need for pre-or post-exposure prophylaxis options. In this context oral delivery would be ideal for use in the early stages of disease management or in a pre-or post-exposure setting. The intrinsically low oral bioavailability of the first-generation 3CLpro inhibitors has been a challenge to their clinical development. Both PF-00835231 and GC-376 have low oral bioavailability in rats of 1.4% and 3%, respectively ( (Boras, Jones et al. 2021) , Patent WO2013049382). The low oral bioavailability and short predicted half-life in human of PF-00835231 was improved with a highly soluble prodrug PF-07304814 that allowed continuous infusion using a minimal dosing volume to reach estimated minimal efficacious levels in clinical trials (Boras, Jones et al. 2021) . The planned route of administration for CDI-45205 is injection or inhalation for potential use as both a therapeutic and prophylactic (https://www.cocrystalpharma.com/news/press-releases/detail/105/cocrystalpharma-selects-lead-compound-for-further). Other less advanced 3CLpro inhibitors have also been administered by subcutaneous or intraperitoneal injection to circumvent the issue of low oral bioavailability. For example, a SARS-CoV-2 3CLpro inhibitor (designated 11a) had a bioavailability of 88% when dosed by intraperitoneal route in mice (Dai, Zhang et al. 2020) . Separate reports that 3C-protease human rhinovirus peptidomimetic inhibitors have oral bioavailability greater than 20% in rodents and other nonclinical species suggest that this chemical class is potentially amenable to oral administration (Dragovich, Prins et al. 2003 , Patick, Brothers et al. 2005 ). At the time of the publishing of this manuscript, Pfizer announced the launch of a Phase 1 clinical study (NCT04756531) with the second-generation orally available 3CLpro inhibitor PF-07321332 (https://cen.acs.org/acs-news/acs-meeting-news/Pfizer-unveils-oral-SARS- CoV/99/i13). PF-07321332 contains a nitrile warhead and was optimized for oral delivery by the reduction of the number of H-bond donors and application of a trifluoroacetyl capping group in P4 (Figure 1 ). The need to discover and develop second-generation 3CLpro inhibitors with improved potency and/or bioavailability is high. In addition, 3CLpro inhibitors commonly target host proteases, which presents a potential liability for unexpected side effects. Therefore, increased target specificity might be critical for future development. However, preclinical evaluation of antiviral potency remains challenging for this class of compounds. Conflicting results of protease inhibitor testing emerged during the early months of the COVID-19 pandemic due to nonstandardized assay conditions among research laboratories. For example, the three drugs shikonin, disulfiram, and ebselen previously approved for other indications were reported to inhibit SARS-CoV-2 3CLpro in enzymatic and antiviral assays (Jin, Du et al. 2020 ). However, enzyme inhibition was only achieved in the absence of physiologically relevant reducing agents, and the reported antiviral effect in cells infected with SARS-CoV-2 could not be reproduced and was therefore suspected to be an indirect consequence of cell death (Gurard-Levin, . Similar lack of reproducibility in SARS-CoV-2 inhibition due to cytotoxicity artefacts have been reported with HIV protease inhibitors nelfinavir and atazanavir (Hattori, Higashi-Kuwata et al. 2021) . These issues highlight the importance of proper assay conditions and controls to ensure correct interpretation of in vitro protease inhibitor testing results. Additional challenges in assessing 3CLpro inhibition potency in infected cells arose from the role of host proteases in virus entry. SARS-CoV-2 uses human ACE2 as its entry receptor and human proteases as entry activators including cell surface transmembrane protease/serine (TMPRSS) proteases, furin, cathepsins, plasmin, elastase, and trypsin (for reviews: (Millet and Whittaker 2015, Luan, Huynh et al. 2020) ). Although the relevance of individual host proteases at the site of infection remains subject to debate, the consensus is that SARS-CoV-2 enters cells mainly through direct membrane fusion by TMPRSS2 protease activation (Hoffmann, Kleine-Weber et al. 2020) . In addition to the TMPRSS2-facilitated direct entry from the cell membrane, SARS-CoV-2 can also enter cells through the endosomal pathway, where spike proteins are proteolytically activated by the lysosomal cathepsin L and/or B proteases (Pislar, Mitrovic et al. J o u r n a l P r e -p r o o f 2020). Therefore, optimizing and developing protease inhibitors for SARS-CoV-2 face distinctive challenges due to the multiple host proteases and their level of redundancy for viral entry. In vitro, the antiviral activity of protease inhibitors largely depends on the mechanism of SARS-CoV-2 cellular entry and is driven by the levels of host protease expression in the cells. For example, the rhinovirus inhibitor rupintrivir and the cathepsin inhibitor K11777 block SARS-CoV-2 replication in A549 lung epithelial cells, but their antiviral effect was greatly diminished when TMRPSS2 was overexpressed (Steuten 2020) . These examples highlight the difficulty of establishing physiologically relevant antiviral assays due to the influence of host protease expression on the mechanism of virus entry. For these reasons, cellular assays risk overpredicting the true potential antiviral effect of broad-spectrum 3CLpro inhibitors that also target cathepsin L and/or other host proteases involved in virus entry . Consequently, it is imperative to validate the protease target (host and/or viral) and the antiviral approach with animal models of virus infection. GC-376 is probably the most studied protease inhibitor in animal models among the current candidates for SARS-CoV-2 treatment. GC-376 administered subcutaneously twice daily resulted in partial to full recovery in laboratory and client-owned cats infected with feline coronavirus (Kim, Lovell et al. 2012 , Pedersen, Kim et al. 2018 . In a mouse model of SARS-CoV-2 infection, intranasal and combined intranasal + intramuscular treatment with GC-376 only achieved marginal reduction in viral load (Shi 2020) . The antiviral effect of GC-376 was improved when combined with GS441524, the parent nucleoside of the remdesivir prodrug. The lack of clear antiviral effect of GC-376 monotherapy in this study might be due to insufficient drug exposure caused by its suboptimal pharmacokinetic properties. Very recently, more convincing animal model efficacy data has emerged with other 3CLpro inhibitors. In a mouse model of SARS-CoV-1 infection, PF-00835231 significantly reduced lung viral titers up to two days post-infection, also alleviated signs of disease such as weight loss and lung pathology (Boras, Jones et al. 2021) . Other molecules derived from boceprevir or telaprevir also demonstrated antiviral activity in a mouse model of SARS-CoV-2 infection (Qiao, Li et al. 2021 ). Finally, ALG-097111 was reported as a potent and highly selective 3CLpro inhibitor with antiviral activity in a SARS-CoV-2 hamster model (Vandyck, Abdelnabi et al. 2021) . The two main classes of direct acting agents for the treatment of SARS-CoV-2 infection are polymerase (nsp12) and protease (3CLpro) inhibitors. The nucleotide analog remdesivir is already approved as a polymerase inhibitor for hospitalized COVID-19 patients, and MK-4482/EIDD-2801, also a polymerase inhibitor, is efficacious in animal models and is currently being evaluated in the clinic (Sheahan, Sims et al. 2020 , Zhu, Chen et al. 2020 , Cox, Wolf et al. 2021 ). In comparison, the most advanced 3CLpro inhibitor PF-07304814 is still in early-stage clinical trial evaluation as an intravenous infusion treatment and has not yet been reported to demonstrate antiviral activity in humans. The emergence of the orally bioavailable clinical candidate PF-07321332 will help to address early-stage non-hospitalized patients as well as potential prophylaxis settings. Although more proof-of-concept work is needed to fully validate 3CLpro inhibitors, this class of compounds provides a promising avenue to treat coronavirus infections either as monotherapies or in combination with other antiviral agents. The authors of this manuscript have the following competing interests: JD is current employee of Aligos Therapeutics, Inc., and KV is current employee of Aligos Belgium BV. The PIKfyve Inhibitor Apilimod: A Double-Edged Sword against COVID-19 Description of the prodrug strategy leading to the increased solubility of PF-07304814 compared to the parent molecule PF-00835231, to allow continuous infusion using a minimal dosing volume to reach estimated minimal efficacious levels in clinical trials The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2 Therapeutically administered ribonucleoside analogue MK-4482/EIDD-2801 blocks SARS-CoV-2 transmission in ferrets Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 8. Pharmacological optimization of orally bioavailable 2-pyridone-containing peptidomimetics The substrate specificity of SARS coronavirus 3C-like proteinase Antiviral Res 182: 104924. *Indicates the importance of reducing agents in 3CLpro assay conditions for inhibitor testing, as well as cytotoxicity as a false positive readout for antiviral effect Coronavirus disease 2019 (COVID-19): A literature review A small molecule compound with an indole moiety inhibits the main protease of SARS-CoV-2 and blocks virus replication Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor This study reveals the importance of TMPRSS2 in SARS-CoV-2 entry Structure of M(pro) from SARS-CoV-2 and discovery of its inhibitors Potent inhibition of enterovirus D68 and human rhinoviruses by dipeptidyl aldehydes and alpha-ketoamides Reversal of the Progression of Fatal Coronavirus Infection in Cats by a Broad-Spectrum Coronavirus Protease Inhibitor Broad-spectrum antivirals against 3C or 3C-like proteases of picornaviruses, noroviruses, and coronaviruses Drug Inhibition of SARS-CoV-2 Replication in Human Pluripotent Stem Cell-Derived Intestinal Organoids Dual Inhibition of SARS-CoV-2 and Human Rhinovirus with Protease Inhibitors in Clinical Development Targeting Proteases for Treating COVID -19 Boceprevir, GC-376, and calpain inhibitors II, XII inhibit SARS-CoV-2 viral replication by targeting the viral main protease Host cell proteases: Critical determinants of coronavirus tropism and pathogenesis In vitro antiviral activity and single-dose pharmacokinetics in humans of a novel, orally bioavailable inhibitor of human rhinovirus 3C protease Efficacy of a 3C-like protease inhibitor in treating various forms of acquired feline infectious peritonitis The role of cysteine peptidases in coronavirus cell entry and replication: The therapeutic potential of cathepsin inhibitors SARS-CoV-2 M(pro) inhibitors with antiviral activity in a transgenic mouse model *Important proof-of-concept demonstration of lung viral load reduction with an oral 3CLpro inhibitor in a transgenic mouse model of SARS-CoV-2 infection 3C-like protease inhibitors block coronavirus replication in vitro and improve survival in MERS-CoV-infected mice An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice The Preclinical Inhibitor GS441524 in Combination with GC376 Efficaciously Inhibited the Proliferation of SARS-CoV-2 in the Mouse Respiratory Tract Challenges for targeting SARS-CoV-2 proteases as a therapeutic strategy for COVID-19 Quantitative electrochemical detection of cathepsin B activity in breast cancer cell lysates using carbon nanofiber nanoelectrode arrays toward identification of cancer formation Structural and inhibitor studies of norovirus 3C-like proteases On the whereabouts of SARS-CoV-2 in the human body: A systematic review ALG-097111, a potent and selective SARS-CoV-2 3-chymotrypsin-like cysteine protease inhibitor exhibits in vivo efficacy in a Syrian Hamster model Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review alpha-Ketoamides as Broad-Spectrum Inhibitors of Coronavirus and Enterovirus Replication: Structure-Based Design, Synthesis, and Activity Assessment New understanding of the damage of SARS-CoV-2 infection outside the respiratory system RNA-Dependent RNA Polymerase as a Target for COVID-19 Drug Discovery We thank Julian Symons and Peggy Korn for their careful editorial review of the manuscript.