Remdesivir treatment methods

Information

  • Patent Grant
  • 11975012
  • Patent Number
    11,975,012
  • Date Filed
    Friday, May 28, 2021
    2 years ago
  • Date Issued
    Tuesday, May 7, 2024
    16 days ago
Abstract
Provided herein are methods of treating or preventing a viral infection in a subject comprising administering a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, wherein the subject is not being treated with chloroquine, or an analog or salt thereof.
Description
BACKGROUND

Preventing or treating some Arenaviridae, Coronaviridae, Filoviridae, Flaviviridae, Orthomyxovirus, Pneumoviridae, and Paramyxoviridae viral infections present challenges due to a lack of vaccine or post-exposure treatment modality for preventing or managing infections caused by viruses from these families. In some cases, patients only receive supportive therapy such as electrolyte and fluid balancing, oxygen, blood pressure maintenance, or treatment for secondary infections.


The compound (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate, referred to herein as the compound of Formula Ia, is known to exhibit antiviral properties against several viral families, including Arenaviridae, Coronaviridae, Filoviridae, Paramyxoviridae, and Flaviviridae viruses (see e.g. Warren, T. et al., Nature (2016) 531:381-385; Lo M K, et al. Sci. Reports 2017; 7:43395; Sheahan T P, et al. Sci. Transl. Med. 2017; 9:eaa13653; Agostini M L, et al. MBio 2018; 9(2):e00221-18; Cell Research (2020) 30:269-271, and WO 2017/184668). There is a need to develop methods of treating viral infections comprising the compound of Formula Ia, or a pharmaceutically acceptable salt thereof.


Methods of treating a viral infection comprising the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human in need thereof should avoid other agents that decrease, retard, or attenuate the antiviral activity of the compound.


BRIEF SUMMARY

In some embodiments, the present disclosure provides a method of treating a viral infection in a human in need thereof, the method comprising administering to the human a therapeutically effective amount of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof:




embedded image



wherein the human is not being treated with chloroquine, or an analog or salt thereof, thereby treating the viral infection.


In some embodiments, the present disclosure provides a method of optimizing a plasma or blood concentration of a compound of Formula II, or a pharmaceutically acceptable salt thereof, in a human in need thereof:




embedded image



the method comprising administering to the human an antiviral compound, wherein the human has not been administered chloroquine, or an analog or salt thereof, the antiviral compound is converted to the compound of Formula II upon administration to the human, and the plasma or blood concentration of the compound of Formula II is optimized in the absence of chloroquine, or an analog or salt thereof.


In some embodiments, the present disclosure provides a method of optimizing a plasma or blood concentration of a compound of Formula II, or a pharmaceutically acceptable salt thereof, in a human in need thereof, the method comprising: (a) administering to the human a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, (b) measuring the plasma or blood concentration of the compound of Formula II in the human; and (c) adjusting any remaining doses of the compound of Formula I, Formula Ia, or Formula Ib, to optimize the plasma or blood concentration of the compound of Formula II in the human.


In some embodiments, the present disclosure provides a method of determining a delivery dose of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, for treating a viral infection in a human in need thereof, the method comprising: (a) providing an original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof; (b) determining whether the human has been administered chloroquine, or an analog or salt thereof; and (c1) if the human has been administered chloroquine, or an analog or salt thereof, increasing the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, to determine the delivery dose, or (c2) if the human has not been administered chloroquine, or an analog or salt thereof, selecting the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, as the delivery dose.


In some embodiments, the present disclosure provides a method of forming a compound of Formula II in a human in need thereof, comprising administering to the human a therapeutically effective amount of a compound of Formula Ia, and instructing the human not to take chloroquine, or an analog or salt thereof, wherein the compound of Formula Ia is metabolized to the compound of Formula II in the absence of chloroquine, or an analog or salt thereof, wherein the compound of Formula II has the structure:




embedded image



and


wherein the compound of Formula Ia has the structure:




embedded image


In some embodiments, the present disclosure provides is a method of reducing the risk of decreased efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection, the method comprising:

  • (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby reducing the risk of decreased efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of preventing a contraindication in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby preventing a contraindication in the human.


In some embodiments, the present disclosure provides a method of maintaining efficacy of a compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby maintaining efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of reducing the risk of a reduced plasma concentration of a compound of Formula II, in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby reducing the risk of a reduced plasma concentration of the compound of Formula II.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Shows the effect of chloroquine (CQ) or hydroxychloroquine (HCQ) on the Formula Ia triphosphate (TP) formation in A549-hACE2 cells.



FIG. 2. Shows the effect of CQ or HCQ on the Formula Ia triphosphate (TP) formation in NHBE cultures.



FIG. 3. Shows the effect of CQ or HCQ on the Formula Ia triphosphate (TP) formation in HEp-2 cells.



FIG. 4A. Shows SARS-CoV-2 antiviral data for the compound of Formula Ia in combination with CQ in A549-hACE2 cells. FIG. 4B. Shows SARS-CoV-2 antiviral data for the compound of Formula Ia in combination with HCQ in A549-hACE2 cells.





DETAILED DESCRIPTION
I. General

The present disclosure provides a method of treating a viral infection in a subject, comprising administering to the subject a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, wherein the subject is not being treated with chloroquine, or an analog or salt thereof.


II. Definitions

A “compound of the disclosure” refers to a compound that is administered to a subject in a method as described herein, and includes compounds of Formula I, Formula Ia, Formula Ib, or a pharmaceutically acceptable salt thereof.


“Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use.


“Pharmaceutically acceptable excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.


“Pharmaceutical composition” refers to a formulation of a compound of the disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, for example, humans. Such a medium includes all pharmaceutically acceptable excipients therefor.


“Effective amount” or “therapeutically effective amount” refers to an amount of a compound of the disclosure, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician. The amount of a compound of the disclosure which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the disclosure, and the age, body weight, general health, sex and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the state of the art, and this disclosure.


“Treatment” or “treating” or “treat” refers to an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results may include one or more of the following: a) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); b) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread (e.g., metastasis) of the disease or condition); and/or c) relieving the disease, that is, causing the regression of clinical symptoms (e.g., ameliorating the disease state, providing partial or total remission of the disease or condition, enhancing effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival.


“Prevention” or “preventing” means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. Compositions may, in some embodiments, be administered to a subject (including a human) who is at risk or has a family history of the disease or condition.


“Subject” or “patient” refer to an animal, such as a mammal, including a human, that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in human therapy and/or veterinary applications. In some embodiments, the subject or the patient is a mammal. In some embodiments, the subject or the patient is human; a domestic animal like a dog or a cat; a farm animal such as a cow, horse, sheep, goat or pig; or a laboratory animal such as a mouse, rat, hamster, guinea pig, pig, rabbit, dog, or monkey. In some embodiments, the subject or the patient is a human.


“Human in need thereof” refers to a human who may have or is suspected to have diseases or conditions that would benefit from certain treatment; for example, being treated with the compounds disclosed herein according to the present application to treat a viral infection.


III. Methods of Use

The present disclosure also provides a method of treating or preventing a viral infection in a human in need thereof, comprising administering a compound described herein.


In some embodiments, the present disclosure provides a method of treating a viral infection in a human in need thereof, the method comprising administering to the human a therapeutically effective amount of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof:




embedded image



wherein the human is not being treated with chloroquine, or an analog or salt thereof, thereby treating the viral infection.


In some embodiments, the present disclosure provides a method of confirming the administration of the compound of Formula I, Formula Ia, or Formula Ib to a human, comprising identifying a compound of Formula II, or a salt thereof, in a biological sample obtained from the human. In some embodiments, the human is not being treated with chloroquine, or an analog or salt thereof. In some embodiments, the human has not been previously treated with chloroquine, or an analog or salt thereof, before the administering of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the biological sample is derived from plasma or blood.


In some embodiments, the present disclosure provides a method of measuring the rate of metabolism of the compound of Formula I, Formula Ia, or Formula Ib in a human, comprising measuring the amount of a compound of Formula II, or a salt thereof, in the human at one or more time points after administration of compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been previously treated with chloroquine, or an analog or salt thereof, before the administering of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a biological sample obtained from the human. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a blood sample. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a plasma sample.


In some embodiments, the present disclosure provides a method of determining the prophylactic or therapeutic response of a human in the treatment of a viral infection comprising measuring the amount of a compound of Formula II, or a salt thereof, in the human at one or more time points after administration of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been previously treated with chloroquine, or an analog or salt thereof, before the administering of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a biological sample obtained from the human. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a blood sample. In some embodiments, the amount of the compound of Formula II, or a salt thereof, is measured from a plasma sample.


A. Chloroquine Administration


The present methods provide treatment of a human that does not have an appreciable systemic concentration of chloroquine, or an analog or salt thereof. In some embodiments, the human in need thereof has not been previously treated with chloroquine, or an analog or salt thereof, before the administering of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, the human has been previously treated with chloroquine, or an analog or salt thereof. In some embodiments, the human in need thereof has not been treated with chloroquine, or an analog or salt thereof, for a period of time before the administering of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure allows for a decrease in the systemic concentration of the chloroquine, or an analog or salt thereof, such that the antiviral activity of the compound of the disclosure is not decreased. For example, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure can allow for a decrease in the plasma concentration of the chloroquine, or an analog or salt thereof, as a result of clearance or metabolism.


In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 14 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 28 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 40 days. In some embodiments, the period of time between the treatment with chloroquine or analog thereof and the administration of the compound of the disclosure is at least 50 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 60 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 90 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 120 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 180 days. In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 365 days.


In some embodiments, the period of time between the treatment with chloroquine, or an analog or salt thereof, and the administration of the compound of the disclosure is at least 30 mins, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 20 hours, or at least 24 hours.


In some embodiments, the human has been administered chloroquine, or an analog or salt thereof, in from about 1 days to about 365 days prior to receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, or from about 1 day to about 14 days, from about 1 day to about 21 days, from about 1 day to about 30 days, from about 1 day to about 45 days, from about 10 days to about 45 days, from about 14 days to about 45 days, from about 21 days to about 45 days, from about 28 days to about 45 days, from about 30 days to about 45 days, 10 days to about 60 days, from about 14 days to about 60 days, from about 21 days to about 60 days, from about 28 days to about 60 days, from about 30 days to about 60 days, from about 40 days to about 60 days, from about 10 days to about 90 days, from about 14 days to about 90 days, from about 21 days to about 90 days, from about 28 days to about 90 days, from about 30 days to about 90 days, from about 40 days to about 90 days, from about 10 days to about 365 days, from about 14 days to about 365 days, from about 21 days to about 365 days, from about 28 days to about 365 days, from about 30 days to about 365 days, or from about 60 days to about 365 days, prior to receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has been administered chloroquine, or an analog or salt thereof, in from about 30 days to about 60 days prior to receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has been administered chloroquine, or an analog or salt thereof, in from about 21 days to about 45 days prior to receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 2 days of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 5 days of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 7 days of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 10 days of receiving the first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 90 days of receiving the first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 365 days of receiving the first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, the method further comprises instructing the human not to take chloroquine, or an analog or salt thereof, during the treatment of the viral infection.


In some embodiments, the method further comprises instructing the human to wait from about 1 day to about 365 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof, or from about 1 day to about 4 days, from about 1 day to about 7 days, from about 1 day to about 10 days, from about 1 day to about 14 days, from about 10 days to about 45 days, from about 14 days to about 45 days, from about 21 days to about 45 days, from about 28 days to about 45 days, from about 30 days to about 45 days, 10 days to about 60 days, from about 14 days to about 60 days, from about 21 days to about 60 days, from about 28 days to about 60 days, from about 30 days to about 60 days, from about 40 days to about 60 days, from about 10 days to about 90 days, from about 14 days to about 90 days, from about 21 days to about 90 days, from about 28 days to about 90 days, from about 30 days to about 90 days, from about 40 days to about 90 days, from about 10 days to about 365 days, from about 14 days to about 365 days, from about 21 days to about 365 days, from about 28 days to about 365 days, from about 30 days to about 365 days, or from about 60 days to about 365 days, after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof.


In some embodiments, the method further comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the method further comprises instructing the human to wait at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof.


In some embodiments, the method further comprises instructing the human to wait from about 30 mins to about one day after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. For example to wait for at least 30 mins, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 20 hours, or at least 24 hours before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof.


In some embodiments, the method further comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the method further comprises instructing the human to wait at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, the method further comprises instructing the human to wait from about 30 mins to about one day after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. For example to wait for at least 30 mins, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 20 hours, or at least 24 hours before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, the human is not administered chloroquine, or an analog or salt thereof, during the treatment of the viral infection. In some embodiments, the method further comprises instructing the human to not administer chloroquine, or an analog or salt thereof, during the treatment of the viral infection.


In some embodiments, the method comprises administering to the human a therapeutically effective amount of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, provided the human has not been administered chloroquine, or an analog or salt thereof, prior to the start of treatment, thereby treating the viral infection. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, for at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days prior to the start of treatment. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, for at least 1 day prior to the start of treatment. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, for at least 10 days prior to the start of treatment. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, for at least 30 mins, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 6 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 20 hours, or at least 24 hours prior to the start of treatment.


Various techniques can be used to determine whether or not a human in need thereof has previously taken chloroquine, or an analog or salt thereof. Non-limiting techniques include self-reporting, interviewing the human, reviewing the human's medical records, or measuring the level of chloroquine, or a metabolite, analog or salt thereof, in the plasma or blood in the human.


The human in need of treatment for a viral infection may also be evaluated for plasma or blood concentration of the chloroquine, or an analog or salt thereof, prior to administration of the compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, a human in need of treatment for a viral infection has a plasma or blood concentration measured prior to administration of the compound of the disclosure.


Concentrations of chloroquine, or an analog or salt thereof, in human plasma or blood can be measured by any method known in the art. See, for example, Walker, O. et al. British Journal Clinical Pharmacology (1983), vol. 16, pages 701-705; Kaewkhao, K. et al. Bioanalysis (2019), vol. 11(5), pages 333-347; Durcan, L. et al. Journal of Rheumatology (2015), vol. 42(11), pages 2092-2097; Munster, T. et al. Arthritis Rheumatology (2002), vol. 46(6), pages 1460-1469.


In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of from about 0.1 ng/mL to about 5000 ng/mL at the time a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered to the human, or from about 0.1 ng/mL to about 4000 ng/mL, from about 0.1 ng/mL to about 3000 ng/mL, from about 0.1 ng/mL to about 2000 ng/mL, from about 0.1 ng/mL to about 1000 ng/mL, from about 0.1 ng/mL to about 500 ng/mL, from about 0.1 ng/mL to about 400 ng/mL, from about 0.1 ng/mL to about 300 ng/mL, from about 0.1 ng/mL to about 200 ng/mL, from about 0.1 ng/mL to about 100 ng/mL, from about 0.1 ng/mL to about 80 ng/mL, from about 0.1 ng/mL to about 60 ng/mL, from about 0.1 ng/mL to about 50 ng/mL, from about 0.1 ng/mL to about 40 ng/mL, from about 0.1 ng/mL to about 30 ng/mL, from about 0.1 ng/mL to about 20 ng/mL, or from about 0.1 ng/mL to about 10 ng/mL, from about 5 ng/mL to about 4000 ng/mL, from about 5 ng/mL to about 3000 ng/mL, from about 5 ng/mL to about 2000 ng/mL, from about 5 ng/mL to about 1000 ng/mL, from about 5 ng/mL to about 500 ng/mL, from about 5 ng/mL to about 400 ng/mL, from about 5 ng/mL to about 300 ng/mL, from about 5 ng/mL to about 200 ng/mL, from about 5 ng/mL to about 100 ng/mL, from about 5 ng/mL to about 80 ng/mL, from about 5 ng/mL to about 60 ng/mL, from about 5 ng/mL to about 50 ng/mL, from about 5 ng/mL to about 40 ng/mL, from about 5 ng/mL to about 30 ng/mL, from about 5 ng/mL to about 20 ng/mL, or from about 5 ng/mL to about 10 ng/mL, at the time a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered to the human. In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of from about 0.1 ng/mL to about 50 ng/mL, at the time a first dose of the compound of the disclosure is administered to the human.


In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 5000 ng/mL at the time a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered to the human, or less than 4000 ng/mL, 3000 ng/mL, 2000 ng/mL, 1000 ng/mL, 500 ng/mL, 400 ng/mL, 300 ng/mL, 200 ng/mL, 100 ng/mL, 80 ng/mL, 60 ng/mL, 50 ng/mL, 45 ng/mL, 40 ng/mL, 35 ng/mL, 30 ng/mL, 25 ng/mL, 20 ng/mL, 15 ng/mL, 10 ng/mL, or 5 ng/mL at the time a first dose of the compound of the disclosure is administered to the human.


In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 50 ng/mL at the time a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered to the human.


“Chloroquine, or an analog or salt thereof” refers to chloroquine (also known as CQ, N′-(7-chloroquinolin-4-yl)-N,N-diethyl-pentane-1,4-diamine and CAS Number 54-05-7), hydroxychloroquine (also known as HCQ, 2-[{4-[(7-chloroquinolin-4-yl)amino]pentyl}(ethyl)amino]ethanol and CAS Number 118-42-3), and metabolites thereof in the plasma after administration to a human. Members include chloroquine, desethylchloroquine (also known as DCQ, 4-N-(7-chloroquinolin-4-yl)-1-N-ethylpentane-1,4-diamine and CAS Number 1476-52-4), hydroxychloroquine, desethylhydroxychloroquine (also known as DHCQ, cletoquine, 2-[4-[(7-chloroquinolin-4-yl)amino]pentylamino]ethanol, and CAS Number 4298-15-1), and bidesethylhydroxychloroquine (also known as BDCQ), or a pharmaceutically acceptable salt thereof. Illustrative examples include chloroquine, or a pharmaceutically acceptable salt thereof. An example is chloroquine phosphate, commercially available as Aralen®. Chloroquine has the chemical structure:




embedded image



An alternative example includes hydroxychloroquine, or a pharmaceutically acceptable salt thereof. An example is hydroxychloroquine sulfate, commercially available as Plaquenil®. Hydroxychloroquine has the chemical structure:




embedded image


In some embodiments, the chloroquine, or an analog or salt thereof, is chloroquine, desethylchloroquine, hydroxychloroquine, desethylhydroxychloroquine, or bidesethylhydroxychloroquine, or a pharmaceutically acceptable salt thereof. In some embodiments, the chloroquine, or an analog or salt thereof, is chloroquine, hydroxychloroquine, or a pharmaceutically acceptable salt thereof. In some embodiments, the chloroquine, or an analog or salt thereof, is chloroquine, or a pharmaceutically acceptable salt thereof. For example, the chloroquine, or an analog or salt thereof, can be chloroquine phosphate. In some embodiments, the chloroquine, or an analog or salt thereof, is hydroxychloroquine, or a pharmaceutically acceptable salt thereof. For example, the chloroquine, or an analog or salt thereof, can be hydroxychloroquine sulfate.


B. Compounds


The present disclosure includes use of antiviral compounds that when administered to a human in need thereof produce the compound of Formula II, or a pharmaceutically acceptable salt thereof.


The present disclosure also includes use of compounds of Formula I, Formula Ia and Formula Ib, or a pharmaceutically acceptable salt thereof.


The compound of Formula I was described in WO2012/012776. The IUPAC name for the compound of Formula I is 2-ethylbutyl ((((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate. The compound of Formula I, or a pharmaceutically acceptable salt thereof, has the structure:




embedded image


The compound of Formula Ia was described in WO2016/069826. The IUPAC name for the compound of Formula Ia is (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyn-olo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate, and the CAS Registry Number is 1809249-37-3. The compound of Formula Ia is also referred to as remdesivir and GS-5734. The compound of Formula Ia, or a pharmaceutically acceptable salt thereof, has the structure:




embedded image


The compound of Formula Ib was described in WO2016/069826. The IUPAC name for the compound of Formula Ib is (S)-2-ethylbutyl 2-(((R)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate. The compound of Formula Ib, or a pharmaceutically acceptable salt thereof, has the structure:




embedded image


In some embodiments, administering a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, to a human in need thereof produces a compound of Formula II or a pharmaceutically acceptable salt thereof:




embedded image


The compound of Formula I, Formula Ia, or Formula Ib can be used in any suitable form. For example, the compound of Formula I, Formula Ia, or Formula Ib can be amorphous or crystalline. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib is amorphous. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib is crystalline.


Crystalline forms of the compound of Formula Ia useful in the methods and compositions of the present disclosure are described in U.S. Patent Application Publication No. 2018/0346504. For example, the compound of Formula Ia can be crystalline Form I, Form II, Form III, or Form IV as described in U.S. Patent Application Publication No. 20180346504, or a combination thereof. In some embodiments, the compound of Formula Ia is crystalline.


The compounds of Formula I, Formula Ia and Formula Ib, or a pharmaceutically acceptable salt thereof, can be combined with one or more pharmaceutically acceptable excipients. In some embodiments, the pharmaceutically acceptable excipient comprises an aqueous vehicle. In some embodiments, the pharmaceutical compositions provided herein comprise the compound of Formula I, or a pharmaceutically acceptable salt thereof, and an aqueous vehicle. In some embodiments, the pharmaceutical compositions provided herein comprise the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and an aqueous vehicle. In some embodiments, the pharmaceutical compositions provided herein comprise the compound of Formula Ib, or a pharmaceutically acceptable salt thereof, and an aqueous vehicle. The aqueous vehicle comprises water and optionally one or more components selected from a co-solvent, a surfactant, a suspending agent, a tonicity agent, a buffer, a cyclodextrin, and an anti-microbial agent or preservative. Exemplary formulations may be found in U.S. Patent Application Publication No. 2019/0083525.


In some embodiments, the method of treating the viral infection in the human in need thereof comprises administering to the human the compound of Formula I, or a pharmaceutically acceptable salt thereof:




embedded image



In some embodiments, the method comprises administering the compound of Formula I.


In some embodiments, the method of treating the viral infection in the human in need thereof, comprises administering to the human a compound of Formula Ia, or a pharmaceutically acceptable salt thereof:




embedded image



In some embodiments, the method comprises administering the compound of Formula Ia.


In some embodiments, the method of treating the viral infection in the human in need thereof comprises administering to the human a compound of Formula Ib, or a pharmaceutically acceptable salt thereof:




embedded image



In some embodiments, the method comprises administering the compound of Formula Ib.


The compound of the disclosure can be administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal (including inhalation), pulmonary, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), implants, and the like. It will be appreciated that the preferred route may vary, for example, with the condition of the human.


In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered by inhalation or intravenously. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered by inhalation.


In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered once daily or twice daily. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered once daily.


In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 4 days. In some embodiments, the second dose of 50-150 mg is administered for an additional 1 to 5 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 9 days. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered once daily. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered over from about 30 to about 120 minutes.


In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 4 days. In some embodiments, the second dose of 100 mg is administered for an additional 1 to 5 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 9 days. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously once daily. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously over from about 30 to about 120 minutes.


In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 4 days. In some embodiments, the second dose of 50-150 mg is administered for an additional 1 to 5 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 150-250 mg on day 1, and administered in a second dose of 50-150 mg on each of the following 9 days. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered once daily. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered over from about 30 to about 120 minutes.


In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 4 days. In some embodiments, the second dose of 100 mg is administered for an additional 1 to 5 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments, the human weighs at least 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 9 days. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously once daily. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously over from about 30 to about 120 minutes.


In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 4 days. In some embodiments, the second dose of 1-5 mg/kg is administered for an additional 1 to 5 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 9 days. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered once daily. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered over from about 30 to about 120 minutes.


In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 4 days. In some embodiments, the second dose of 2.5 mg/kg is administered for an additional 1 to 5 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 9 days. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously once daily. In some embodiments, the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously over from about 30 to about 120 minutes.


In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 4 days. In some embodiments, the second dose of 1-5 mg/kg is administered for an additional 1 to 5 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered in the first dose of 2.5-10 mg/kg on day 1, and administered in a second dose of 1-5 mg/kg on each of the following 9 days. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered once daily. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered over from about 30 to about 120 minutes.


In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 4 days. In some embodiments, the second dose of 2.5 mg/kg is administered for an additional 1 to 5 days. In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 4, 5, 6, 7, 8, or 9 days. In some embodiments, the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 9 days. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously once daily. In some embodiments, the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, is administered intravenously over from about 30 to about 120 minutes.


C. Viral Infections


Any suitable viral infection can be treated by the method of the present disclosure. In some embodiments, the viral infection is caused by a virus selected from the group consisting of Arenaviridae, Coronaviridae, Filoviridae, Flaviviridae, Pneumoviridae, and Paramyxoviridae.


In some embodiments, the viral infection is caused by an Arenaviridae virus. In some embodiments, the method of treating an Arenaviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the method comprises treating an Arenaviridae virus infection selected from the group consisting of Allpahuayo virus (ALLV), Amapari virus (AMAV), Bear Canyon virus (BCNV), Catarina virus, Chapare virus, Cupixi virus (CPXV), Dandenong virus, Flexal virus (FLEV), Guanarito virus (GTOV), Ippy virus (IPPYV), Junin virus (JUNV), Kodoko virus, Lassa virus (LASV), Latino virus (LATV), Lymphocytic choriomeningitis virus (LCMV), Lujo virus, Machupo virus (MACV), Mobala virus (MOBV), Morogoro virus, Mopeia virus (MOPV), Oliveros virus (OLVV), Parana virus (PARV), Pichinde virus (PICV), Pinhal virus, Pirital virus (PIRV), Sabia virus (SABV), Skinner Tank virus, Tacaribe virus (TCRV), Tamiami virus (TAMV), and Whitewater Arroyo virus (WWAV) by administering a compound of the disclosure provided herein. In some embodiments, the Arenaviridae virus is Lassa or Junin. In some embodiments, the method comprises treating a Lassa virus infection by administering a compound of the disclosure provided herein. In some embodiments, the method comprises treating a Junin virus infection by administering a compound of the disclosure provided herein.


In some embodiments, the viral infection is caused by a Coronaviridae virus. In some embodiments, the method of treating a Coronaviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the Coronaviridae virus infection is selected from the group consisting of a Severe Acute Respiratory Syndrome (SARS) infection, SARS-CoV-2 (also known as 2019-nCov and COVID-19) infection, Middle Eastern Respiratory Syndrome (MERS) infection, other human coronavirus (229E, NL63, 0C43, HKU1, or WW1) infections, or a zoonotic coronavirus (PEDV or HKU CoV isolates such as HKU3, HKU5, or HKU9) infection. In some embodiments, the Coronaviridae virus is SARS, SARS-CoV-2, or MERS. In some embodiments, the Coronaviridae virus is SARS. In some embodiments, the Coronaviridae virus is SARS-CoV-2. In some embodiments, the Coronaviridae virus is MERS. In some embodiments, the viral infection is caused by a virus having at least 70% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase. In some embodiments, the viral infection is caused by a virus having at least 80% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase. In some embodiments, the viral infection is caused by a virus having at least 90% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase. In some embodiments, the viral infection is caused by a virus having at least 95% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase. In some embodiments, the viral infection is caused by a virus having at least 97% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase. In some embodiments, the viral infection is caused by a virus having at least 99% sequence homology a viral polymerase selected from SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase.


In some embodiments, the viral infection is caused by a Filoviridae virus. In some embodiments, the method of treating a Filoviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the Filoviridae virus is Ebola or Marburg. In some embodiments, the Filoviridae virus is an Ebola virus. In some embodiments, the Ebola virus is selected from the group consisting of: Zaire (i.e. Ebola virus, EBOV), Sudan, Tai Forest, Bundibugyo, and Reston. In some embodiments, the Filoviridae virus is a Marburg virus.


In some embodiments, the viral infection is caused by a Flaviviridae virus. In some embodiments, the method of treating a Flaviviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the Flaviviridae virus is selected from the group consisting of: dengue, yellow fever, West Nile, and Zika. In some embodiments, the method of treating a dengue virus infection comprises administering a compound of the disclosure provided herein. In some embodiments, the Flaviviridae virus is yellow fever. In some embodiments, the method of treating a yellow fever virus infection comprises administering a compound of the disclosure provided herein. In some embodiments, the method of treating a West Nile virus infection comprises administering a compound of the disclosure provided herein. In some embodiments, the method of treating a Zika virus infection comprises administering a compound of the disclosure provided herein. In some embodiments, the method of treating a hepatitis C virus infection comprises administering a compound of the disclosure provided herein.


In some embodiments, the viral infection is caused by a Pneumoviridae virus. In some embodiments, the method of treating a Pneumoviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the Pneumoviridae virus is respiratory syncytial virus or human metapneumovirus. In some embodiments, the Pneumoviridae virus is respiratory syncytial virus. In some embodiments, the Pneumoviridae virus is human metapneumovirus.


In some embodiments, the viral infection is caused by a Paramyxoviridae virus. In some embodiments, the method of treating a Paramyxoviridae virus infection comprises administering a compound of the disclosure, such as a compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof. Paramyxoviridae viruses include, but are not limited to Nipah virus, Hendra virus, measles, mumps, and parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah or parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah. In some embodiments, the Paramyxoviridae virus is parainfluenza.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating an Arenaviridae virus infection. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating an Arenaviridae virus infection selected from the group of Allpahuayo virus (ALLY), Amapari virus (AMAV), Bear Canyon virus (BCNV), Catarina virus, Chapare virus, Cupixi virus (CPXV), Dandenong virus, Flexal virus (FLEV), Guanarito virus (GTOV), Ippy virus (IPPYV), Junin virus (JUNV), Kodoko virus, Lassa virus (LASV), Latino virus (LATV), Lymphocytic choriomeningitis virus (LCMV), Lujo virus, Machupo virus (MACV), Mobala virus (MOBV), Morogoro virus, Mopeia virus (MOPV), Oliveros virus (OLVV), Parana virus (PARV), Pichinde virus (PICV), Pinhal virus, Pirital virus (PIRV), Sabia virus (SABV), Skinner Tank virus, Tacaribe virus (TCRV), Tamiami virus (TAMV), and Whitewater Arroyo virus (WWAV) by administering a compound of the disclosure provided herein. In some embodiments, the Arenaviridae virus is Lassa or Junin. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating a Lassa virus infection. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating a Junin virus infection.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein for use in methods of treating a Coronaviridae virus infection. In some embodiments, the Coronaviridae virus infection is selected from the group consisting of a Severe Acute Respiratory Syndrome (SARS) infection, SARS-CoV-2 (also known as 2019-nCov and COVID-19) infection, Middle Eastern Respiratory Syndrome (MERS) infection, other human coronavirus (229E, NL63, 0C43, HKU1, or WW1) infections, or a zoonotic coronavirus (PEDV or HKU CoV isolates such as HKU3, HKU5, or HKU9) infection. In some embodiments, the Coronaviridae virus is SARS, SARS-CoV-2, or MERS. In some embodiments, the Coronaviridae virus is SARS. In some embodiments, the present disclosure provides a compound of the disclosure provided herein for use in methods of treating a Severe Acute Respiratory Syndrome (SARS) infection. In some embodiments, the Coronaviridae virus is SARS-CoV-2. In some embodiments, the present disclosure provides a compound of the disclosure provided herein for use in methods of treating a SARS-nCoV-2 infection. In some embodiments, the Coronaviridae virus is MERS. In some embodiments, the present disclosure provides a compound of the disclosure provided herein for use in methods of treating a Middle East Respiratory Syndrome (MERS) infection.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating a Filoviridae virus infection. In some embodiments, the Filoviridae virus is Ebola or Marburg. In some embodiments, the Filoviridae virus is an Ebola virus. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating an Ebola virus infection. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating an Ebola virus infection selected from the group consisting of: Zaire (i.e. Ebola virus, EBOV), Sudan, Tai Forest, Bundibugyo, and Reston. In some embodiments, the Filoviridae virus is a Marburg virus. In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating a Marburg virus infection.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in methods of treating a Flaviviridae virus infection. In some embodiments, the Flaviviridae virus is selected from the group consisting of: dengue, yellow fever, West Nile, and Zika. In some embodiments, the Flaviviridae virus is dengue virus. In some embodiments, the Flaviviridae virus is yellow fever. In some embodiments, the Flaviviridae virus is West Nile virus. In some embodiments, the Flaviviridae virus is Zika virus. In some embodiments, the Flaviviridae virus is hepatitis C virus.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in a method of treating a Pneumoviridae virus infection. In some embodiments, the Pneumoviridae virus is respiratory syncytial virus or human metapneumovirus. In some embodiments, the Pneumoviridae virus is a respiratory syncytial virus. In some embodiments, the Pneumoviridae virus is human metapneumovirus.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in a method of treating a Paramyxoviridae virus infection. Paramyxoviridae viruses include, but are not limited to Nipah virus, Hendra virus, measles, mumps, and parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah or parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah. In some embodiments, the Paramyxoviridae virus is parainfluenza.


In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an Arenaviridae virus infection. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an Arenaviridae virus infection selected from the group of: Allpahuayo virus (ALLY), Amapari virus (AMAV), Bear Canyon virus (BCNV), Catarina virus, Chapare virus, Cupixi virus (CPXV), Dandenong virus, Flexal virus (FLEV), Guanarito virus (GTOV), Ippy virus (IPPYV), Junin virus (JUNV), Kodoko virus, Lassa virus (LASV), Latino virus (LATV), Lymphocytic choriomeningitis virus (LCMV), Lujo virus, Machupo virus (MACV), Mobala virus (MOBV), Morogoro virus, Mopeia virus (MOPV), Oliveros virus (OLVV), Parana virus (PARV), Pichinde virus (PICV), Pinhal virus, Pirital virus (PIRV), Sabia virus (SABV), Skinner Tank virus, Tacaribe virus (TCRV), Tamiami virus (TAMV), and Whitewater Arroyo virus (WWAV). In some embodiments, the Arenaviridae virus is Lassa or Junin. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Lassa virus infection. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Junin virus infection.


In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Coronaviridae virus infection. In some embodiments, the Coronaviridae virus infection is selected from the group consisting of a Severe Acute Respiratory Syndrome (SARS) infection, SARS-CoV-2 (also known as 2019-nCov and COVID-19) infection, Middle Eastern Respiratory Syndrome (MERS) infection, other human coronavirus (229E, NL63, 0C43, HKU1, or WW1) infections, or a zoonotic coronavirus (PEDV or HKU CoV isolates such as HKU3, HKU5, or HKU9) infection. In some embodiments, the Coronaviridae virus is SARS, SARS-CoV-2, or MERS. In some embodiments, the Coronaviridae virus is SARS. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a SARS infection. In some embodiments, the Coronaviridae virus is SARS-CoV-2. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a SARS-nCoV-2 infection. In some embodiments, the Coronaviridae virus is MERS. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a MERS infection.


In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Filoviridae virus infection. In some embodiments, the Filoviridae virus is Ebola or Marburg. In some embodiments, the Filoviridae virus is an Ebola virus. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an Ebola virus infection. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating an Ebola virus infection selected from the group consisting of: Zaire (i.e. Ebola virus, EBOV), Sudan, Tai Forest, Bundibugyo, and Reston. In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Marburg virus infection.


In some embodiments, the present disclosure provides the use of a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a Flaviviridae virus infection. In some embodiments, the Flaviviridae virus is selected from the group consisting of: dengue, yellow fever, West Nile, and Zika. In some embodiments, the Flaviviridae virus is dengue virus. In some embodiments, the Flaviviridae virus is yellow fever. In some embodiments, the Flaviviridae virus is West Nile virus. In some embodiments, the Flaviviridae virus is Zika virus. In some embodiments, the Flaviviridae virus is hepatitis C virus.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in a manufacture of a medicament for treating a Pneumoviridae virus infection. In some embodiments, the Pneumoviridae virus is respiratory syncytial virus or human metapneumovirus. In some embodiments, the Pneumoviridae virus is a respiratory syncytial virus. In some embodiments, the Pneumoviridae virus is human metapneumovirus.


In some embodiments, the present disclosure provides a compound of the disclosure provided herein, or pharmaceutically acceptable salt thereof, for use in a manufacture of a medicament for treating a Paramyxoviridae virus infection. Paramyxoviridae viruses include, but are not limited to Nipah virus, Hendra virus, measles, mumps, and parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah or parainfluenza virus. In some embodiments, the Paramyxoviridae virus is Nipah. In some embodiments, the Paramyxoviridae virus is parainfluenza.


D. Additional Uses


In some embodiments, the present disclosure provides a method of optimizing a plasma or blood concentration of a compound of Formula II, or a pharmaceutically acceptable salt thereof, in a human in need thereof:




embedded image



the method comprising administering to the human an antiviral compound, wherein the human has not been administered chloroquine, or an analog or salt thereof, the antiviral compound is converted to the compound of Formula II upon administration to the human, and the plasma or blood concentration of the compound of Formula II is optimized in the absence of chloroquine, or an analog or salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day of administering to the human an antiviral compound. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 10 days of administering to the human an antiviral compound.


In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 mins, 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 20 hours, or 24 hours of administering to the human an antiviral compound. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days, of administering to the human an antiviral compound. In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 50 ng/mL, such as less than 45 ng/mL, 40 ng/mL, 35 ng/mL, 30 ng/mL, 25 ng/mL, 20 ng/mL, 15 ng/mL, 10 ng/mL, or 5 ng/mL, at the time a first dose of the antiviral compound is administered to the human. In some embodiments, the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 50 ng/mL, at the time a first dose of the antiviral compound is administered to the human.


In some embodiments, the antiviral compound is a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the plasma or blood concentration of the compound of Formula II in the human is higher than a second concentration of a compound of Formula II in a reference human treated with chloroquine, or an analog or salt thereof, and the antiviral compound. In some embodiments, the plasma or blood concentration of the compound of Formula II in the human is from about 1.1 times to about 10 times higher, such as from about 1.2 times to about 5 times, from about 1.3 times to about 5 times, from about 1.2 times to about 4 times, from about 1.3 times to about 4 times, from about 1.2 times to about 3 times, from about 1.3 times to about 3 times, from about 1.2 times to about 2 times, or from about 1.3 times to about 2 times, higher than a second concentration of the compound in a reference human treated with chloroquine, or an analog or salt thereof, and the antiviral compound. In some embodiments, the plasma or blood concentration of the compound of Formula II in the human is at least 1.1 times higher, such as at least 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3 times, 3.2 times, 3.5 times, 3.6 times, 3.8 times, 4 times, 4.2 times, 4.4 times, 4.6 times, 4.8 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times higher than a second concentration of the compound in a reference human treated with chloroquine, or an analog or salt thereof, and the antiviral compound.


In some embodiments, the present disclosure provides a method of optimizing a plasma or blood concentration of a compound of Formula II, or a pharmaceutically acceptable salt thereof, in a human in need thereof, comprises: (a) administering to the human a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof (b) measuring the plasma or blood concentration of the compound of Formula II in the human; and (c) adjusting any remaining doses of the compound of Formula I, Formula Ia, or Formula Ib, to optimize the plasma or blood concentration of the compound of Formula II in the human. In some embodiments, the method comprises administering to the human a daily dose. In some embodiments, the method comprises administering to the human 10 daily doses.


In some embodiments, the plasma or blood concentration of the compound of Formula II in the human is from about 1.1 times to about 10 times higher, such as from about 1.2 times to about 5 times, from about 1.3 times to about 5 times, from about 1.2 times to about 4 times, from about 1.3 times to about 4 times, from about 1.2 times to about 3 times, from about 1.3 times to about 3 times, from about 1.2 times to about 2 times, or from about 1.3 times to about 2 times, higher than a second concentration of the compound in a reference human treated with chloroquine, or an analog or salt thereof, and a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the plasma or blood concentration of the compound of Formula II in the human is at least 1.1 times higher, such as at least 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3 times, 3.2 times, 3.5 times, 3.6 times, 3.8 times, 4 times, 4.2 times, 4.4 times, 4.6 times, 4.8 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times higher than a second concentration of the compound in a reference human treated with chloroquine, or an analog or salt thereof, and a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of determining a delivery dose of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, for treating a viral infection in a human in need thereof, the method comprising: (a) providing an original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof; (b) determining whether the human has been administered chloroquine, or an analog or salt thereof; and (c1) if the human has been administered chloroquine, or an analog or salt thereof, increasing the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, to determine the delivery dose, or (c2) if the human has not been administered chloroquine, or an analog or salt thereof, selecting the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, as the delivery dose.


In some embodiments, the method of determining a delivery dose comprises determining whether the human has been administered chloroquine, or an analog thereof, within 1 day prior to the treatment of the viral infection. In some embodiments, the method comprises determining whether the human has been administered chloroquine, or an analog thereof, within 10 days prior to the treatment of the viral infection. In some embodiments, determining whether the human has been administered chloroquine, or an analog thereof, comprises self-reporting, interviewing the human, reviewing the human's medical records, or measuring the level of chloroquine, or an analog or salt thereof, in the plasma or blood in the human. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days, of administering to the human an antiviral compound.


In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof.


In some embodiments, when the human has been administered chloroquine, or an analog or salt thereof, prior to treating the viral infection, the method comprises increasing the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof, by from about 1.1 times to about 10 times higher, to determine the delivery dose, or from about 1.2 times to about 5 times, from about 1.3 times to about 5 times, from about 1.2 times to about 4 times, from about 1.3 times to about 4 times, from about 1.2 times to about 3 times, from about 1.3 times to about 3 times, from about 1.2 times to about 2 times, or from about 1.3 times to about 2 times, higher to determine the delivery dose. In some embodiments, when the human has been administered chloroquine, or an analog or salt thereof, prior to treating the viral infection, the method comprises increasing the original dose of the compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof, by at least 1.1 times higher to determine the delivery dose, or at least 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3 times, 3.2 times, 3.5 times, 3.6 times, 3.8 times, 4 times, 4.2 times, 4.4 times, 4.6 times, 4.8 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times higher, to determine the delivery dose. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days, of administering to the human an antiviral compound.


In some embodiments, the present disclosure provides a method of forming a compound of Formula II in a human in need thereof, comprising administering to the human a therapeutically effective amount of a compound of Formula Ia, and instructing the human not to take chloroquine, or an analog or salt thereof, wherein the compound of Formula Ia is metabolized to the compound of Formula II in the absence of chloroquine, or an analog or salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours of receiving a first dose of the compound of Formula Ia. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days, of administering the compound of Formula Ia. In some embodiments, after instructing the human not to take chloroquine, or an analog or salt thereof, the human waits at least 1 day before administering the compound of Formula Ia.


In some embodiments, the present disclosure provides a method of reducing the risk of decreased efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection, comprises: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby reducing the risk of decreased efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human has not been administered chloroquine, or an analog or salt thereof, within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days, of administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, if the human is determined to have taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, the human waits at least 1 day before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


For the methods of the present disclosure requiring instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, any number of actions alone or in combination can be taken in the event that the human is administered chloroquine, or an analog or salt thereof, after receiving the instructions and after receiving the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, the human stops taking chloroquine, or an analog or salt thereof. In some embodiments, the human waits after the last dose of chloroquine, or an analog or salt thereof, for at least 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours before receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, the human waits after the last dose of chloroquine, or an analog or salt thereof, for at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days before receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof. In some embodiments, after instructing the human not to take chloroquine, or an analog or salt thereof, the human waits at least 1 day before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the human takes an additional 1 dose, 2 doses, 3 doses, 4 doses, 5 doses, 6 doses, 7 doses, 8 doses, 9 doses, or 10 doses of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, as compared to a reference human who has not been administered chloroquine, or an analog or salt thereof. In some embodiments, the human takes a higher dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, as compared to a reference human who has not been administered chloroquine, or an analog or salt thereof, or at least 1.1 times, 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3 times, 3.2 times, 3.5 times, 3.6 times, 3.8 times, 4 times, 4.2 times, 4.4 times, 4.6 times, 4.8 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times higher dose of the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, as compared to a reference human who has not been administered chloroquine, or an analog or salt thereof.


In some embodiments, the present disclosure provides a method of preventing a contraindication in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby preventing a contraindication in the human. In some embodiments, if the human is determined to have taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, the human waits at least 1 day before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, preventing a contraindication in the human is in comparison to a second human who has taken chloroquine, or an analog or salt thereof, prior to the administration of the compound of Formula Ia, or pharmaceutically acceptable salt thereof, or who has not been instructed not to take chloroquine, or an analog or salt thereof. In some embodiments, the second human who has taken chloroquine, or an analog or salt thereof, prior to the administration of the compound of Formula Ia, or pharmaceutically acceptable salt thereof, is more likely than the human to have a contraindication after administration of the compound of Formula Ia or pharmaceutically acceptable salt thereof. In some embodiments, the second human who has not been instructed not to take chloroquine, or an analog or salt thereof, prior to the administration of the compound of Formula Ia, or pharmaceutically acceptable salt thereof, is more likely than the human to have a contraindication after administration of the compound of Formula Ia or pharmaceutically acceptable salt thereof. In some embodiments, the second human is at least 1.1 times, such as 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3 times, 3.2 times, 3.5 times, 3.6 times, 3.8 times, 4 times, 4.2 times, 4.4 times, 4.6 times, 4.8 times, 5 times, 6 times, 7 times, 8 times, 9 times, or 10 times more likely than the human to have a contraindication after administration of the compound of Formula Ia or pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of maintaining efficacy of a compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby maintaining efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, maintaining efficacy comprises maintaining a sufficient plasma or blood concentration of the compound of Formula II in the human after administration as compared to a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, the plasma or blood concentration of the compound of Formula II in the human after administration is from about 50% to about 100% of a second plasma or blood concentration of the compound of Formula II in a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, or from about 60% to about 100%, from about 70% to about 100%, from about 80% to about 100%, or from about 90% to about 100%, of a second plasma or blood concentration of the compound of Formula II in a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, if the human is determined to have taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, the human waits at least 1 day before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of reducing the risk of a reduced plasma concentration of a compound of Formula II, in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, (b) instructing the human not to take chloroquine, or an analog or salt thereof, while being treated with the compound a of Formula Ia, or a pharmaceutically acceptable salt thereof, and (c) administering the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, to the human, thereby reducing the risk of a reduced plasma concentration of the compound of Formula II. In some embodiments, if the human is determined to have taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, the human waits at least 1 day before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, reducing the risk of a reduced plasma concentration of a compound of Formula II comprises maintaining a sufficient concentration of the compound of Formula II in the plasma of the human after administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, as compared to a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof. In some embodiments, the plasma concentration of the compound of Formula II in the human after administration is from about 50% to about 100% of a second concentration of the compound of Formula II in a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, or from about 60% to about 100%, from about 70% to about 100%, from about 80% to about 100%, or from about 90% to about 100%, of a second concentration of the compound of Formula II in a second human that has not taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof.


For the methods of the present disclosure requiring determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, the determining can be performed by any suitable means. For example, non-limiting techniques include self-reporting, interviewing the human, reviewing the human's medical records, or measuring the level of chloroquine, or an analog or salt thereof, in the plasma or blood in the human. In some embodiments, the determining includes measuring the level of chloroquine, or a metabolite, analog or salt thereof, in the plasma or blood in the human.


IV. Kits

In some embodiments, the present disclosure provides the use of a kit comprising a compound or composition disclosed herein. In some embodiments, the kit further comprises a label and/or instructions for using the compound or pharmaceutical composition in the method of the present disclosure. For instance, in some embodiments, the kit comprises instructing the human not to take chloroquine, or an analog or salt thereof, during the treatment of the viral infection.


In some embodiments, the kit comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, at least 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, such as at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 1 day after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 10 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 14 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 21 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula I, Formula Ia or Formula Ib, or pharmaceutically acceptable salt thereof.


In some embodiments, the kit comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, at least 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, or 20 hours before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait after taking chloroquine, or an analog or salt thereof, such as at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 14 days, 21 days, 28 days, 30 days, 45 days, 60 days, 75 days, 90 days, 120 days, 150 days, 180 days, 210 days, 240 days, 270 days, 300 days, 330 days, or 365 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 10 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 14 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises instructing the human to wait at least 21 days after taking chloroquine, or an analog or salt thereof, before administering the compound of Formula Ia, or pharmaceutically acceptable salt thereof.


In some embodiments, the kit further comprises a nebulizer. Any suitable nebulizer may be used. In some embodiments, the nebulizer is a glass nebulizer. In some embodiments, the nebulizer is a hand bulb nebulizer. In some embodiments, the nebulizer is a jet nebulizer or a vibrating mesh nebulizer. In some embodiments, the nebulizer is a jet nebulizer (e.g. VixOne™ AeroEclipse®, Pari LC® Plus). In some examples the nebulizer is a vibrating mesh nebulizer (e.g. eFlow® rapid). In some embodiments, the nebulizer is a ultrasonic nebulizer. In some embodiments, the nebulizer is an adaptive aerosol delivery nebulizer. In some embodiments, the nebulizer is a metered dose inhaler (e.g. a metered dose liquid inhaler).


V. Examples
Example 1. Chloroquine Effect on Compound of Formula Ia in Cells

Antiviral potency of the compound of Formula Ia against respiratory syncytial virus (RSV) was determined in HEp2 cells in the following manner. HEp2 cells (3×103/well) were suspended in DMEM medium+GlutaMAX (supplemented with 10% FBS and 1% Penicillin/Streptomycin) and seeded into 96 well plates. After a 4-hour incubation at 37° C.+5% CO2, three-fold serial dilutions of the compound (final RDV concentration of 9.28 nM to 2000 nM) were added to each well using an HP D300e digital dispenser. To determine the effect of chloroquine (CQ) on Formula Ia antiviral activity, the Formula Ia dilution series was additionally spiked with DMSO or 4× increasing concentrations of CQ (final concentrations: 10 nM, 40 nM, 160 nM, 640 nM, and 2560 nM). The cells were then infected with RSV A2 virus diluted in DMEM medium+GlutaMAX at an MOI=4 and incubated for 4 days at 37° C. and 5% CO2. The final volume in each well was 200 μL. Uninfected and untreated wells were included as controls for 100% cell viability. Following the incubation, 100 μL of culture supernatant was removed from each well and replaced with 100 μL of CellTiter-Glo reagent (Promega). The plates were then rocked for 2 minutes followed by a 10-minute incubation at 25° C. Cell viability was then assessed by measuring luminescence signal using and Envision plate reader. Values were normalized to the uninfected and infected DMSO controls (as 100% and 0% infection, respectively) and data was fit using non-linear regression analysis with the XLfit software. EC50 values were determined as the point the non-linear regression curve of 50% infection. The compiled data was generated using three biological replicates each containing technical duplicates for each CQ concentration.


The effect of the combination of the compound of Formula Ia and concentrations of chloroquine (CQ) on RSV replication in HEp2 cells were measured. Results show that CQ antagonizes the anti-RSV activity of the compound of Formula Iain a dose-dependent manner as demonstrated by higher Formula Ia EC50 values with increasing concentrations of CQ (Table 1).









TABLE 1







Effect of Chloroquine on Anti-RSV Activity of Formula Ia in HEp2 Cells










Chloroquine
Formula Ia RSV
Formula Ia RSV



(nM)
EC50 (nM)
EC50 fold change
P value













0
 49.5 ± 13.1
1



10
 58.4 ± 12.8
1.19 ± 0.10
0.075


40
 72.1 ± 24.8
1.45 ± 0.19
0.052


160
 99.6 ± 30.3
2.02 ± 0.17
0.0088


640
138.3 ± 33.8
2.86 ± 0.50
0.023


2560
209.0 ± 18.2
4.43 ± 1.13
0.034









Example 2: Metabolism Data

A549-hACE2 and NHBE cells were seeded in 6-well plates at 4×105 and 2.5×105 cells/well, respectively. HEp-2 cells were seeded in 12-well plates at 2.5×105 cells/well. Twenty-four hours later, cell culture medium was replaced with medium containing 1 μM Formula Ia alone, or in combination with 1 or 10 μM of either CQ or HCQ, and incubated at 37° C. At 2, 8, 24 and 48 h post drug addition, cells were washed 3 times with ice-cold tris-buffered saline, scraped into 0.5 mL ice-cold 70% methanol and stored at −80° C. Extracts were centrifuged at 15,000 g for 15 minutes and supernatants were transferred to clean tubes for evaporation in a miVac Duo concentrator (Genevac). Dried samples were reconstituted in mobile phase A containing 3 mM ammonium formate (pH 5) with 10 mM dimethylhexylamine (DMHA) in water for analysis by LC-MS/MS, using a multi-stage linear gradient from 10% to 50% acetonitrile in mobile phase A at a flow rate of 360 μL/min. Analytes were separated using a 50×2 mm, 2.5 μm Luna C18(2) HST column (Phenomenex) connected to an LC-20ADXR (Shimadzu) ternary pump system and an HTS PAL autosampler (LEAP Technologies). Detection was performed on a Qtrap (6500+AB Sciex) mass spectrometer operating in positive ion and multiple reaction monitoring modes. Analytes were quantified using a 7-point standard curve ranging from 0.624 to 160 pmol per million cells prepared in extracts from untreated cells. For normalization by cell number, multiple untreated culture wells were counted at each timepoint.


The metabolism of the compound of Formula Ia to the active triphosphate species (Formula Ia-TP) species was evaluated in A549-hACE2, NHBE, and HEp-2 cell cultures following incubation with the compound of Formula Ia alone and when combined with CQ or HCQ. The concentrations of CQ and HCQ used in these in vitro assays correspond to systemic and lung exposures that result from doses recommended under the former EUA guidance for COVID-19. The average systemic exposures range from 610-760 nM for CQ and approximately 1.7 μM for HCQ, which can achieve lung concentrations in excess of 20 μM for either drug {Salman 2017, Zhao 2014}. In A549-hACE2 cells, co-incubation of the compound of Formula Ia and 1 μM CQ or HCQ did not significantly reduce Formula Ia-TP levels compared to the Formula Ia-TP in the absence of CQ or HCQ; however, incubation at 10 μM of either CQ or HCQ did significantly lower the production of Formula Ia-TP (Table 2; FIG. 1). While Formula Ia-TP levels trended lower in NHBE cultures treated with either concentration of CQ or 1 μM HCQ, these differences were not statistically significant. Conversely, treatment of NHBE cultures with 10 μM HCQ significantly reduced Formula Ia-TP levels (Table 3; FIG. 2). In HEp-2 cervical carcinoma cells, treatment with CQ or HCQ at either 1 or 10 μM showed a dose-dependent reduction in formation of intracellular Formula Ia-TP (Table 4; FIG. 3). Significant CQ or HCQ antagonism of Formula Ia-TP formation in Hep-2 cells was observed at 8 h post treatment and persisted for 48 h. The observed reductions in Formula Ia-TP in the presence of CQ or HCQ indicates a potential antagonistic effect on the compound of Formula Ia metabolism to its active triphosphate.









TABLE 2







Effect of CQ or HCQ on RDV-TP Formation in A549-hACE2 Cells









Mean RDV-TP (pmol/million cells)a














1 μM
1 μM
1 μM
1 μM Formula




Formula Ia
Formula Ia
Formula Ia
Ia


Treatment
1 μM
+
+
+
+


time (h)
Formula Ia
1 μM CQ
10 μM CQ
1 μM HCQ
10 μM HCQ





 2
3.6 ± 0.2
2.4 ± 0.1
0.7 ± 0.1
2.8 ± 0.3
1.0 ± 0.1


 8
11.4 ± 0.8 
10.0 ± 0.3 
2.5 ± 0.1
10.3 ± 1.2 
2.2 ± 0.3


24
6.8 ± 1.3
5.8 ± 1.2
1.7 ± 0.4
7.0 ± 2.0
2.2 ± 0.6


48
3.0 ± 0.5
2.9 ± 0.6
1.8 ± 0.4
2.8 ± 0.3
1.8 ± 0.4


Average
6.5 ± 0.8
5.6 ± 0.8
1.8 ± 0.3
6.2 ± 1.3
2.0 ± 0.4


Formula Ia-TP







concentration







(pmol/million cells)b







P valuec
NA
0.8497
0.0030
0.9960
0.0041






aValues are mean ± SEM of duplicate samples collected at each timepoint from two separate experiments.




bAverage Formula Ia-TP concentration was determined using GraphPad Prism 8.1.2 from the total area under the curve from Formula Ia-TP concentrations detected by LC-MS at 0, 2, 8, 24, and 48 h; and divided by the total time of the assay (48 h).




cP value is determined using one-way ANOVA with Dunnett's multiple comparison analysis of Formula Ia + DMSO compared to various concentrations of CQ or HCQ using GraphPad Prism 8.1.2.














TABLE 3







Effect of CQ or HCQ on RDV-TP Formation in NHBE Cultures









Mean RDV-TP (pmol/million cells)a














1 μM
1 μM
1 μM
1 μM Formula




Formula Ia
Formula Ia
Formula Ia
Ia


Treatment
1 μM
+
+
+
+


time (h)
Formula Ia
1 μM CQ
10 μM CQ
1 μM HCQ
10 μM HCQ





 2
4.4 ± 0.5
2.2 ± 0.2
1.8 ± 0.5
2.2 ± 0.2
1.1 ± 0.5


 8
19.4 ± 3.8 
12.1 ± 2.8 
12.3 ± 4.6 
11.1 ± 1.8 
9.9 ± 1.4


24
16.7 ± 1.0 
12.9 ± 1.2 
9.7 ± 0.6
10.8 ± 1.2 
9.2 ± 1.2


48
14.2 ± 0.6 
14.7 ± 0.5 
13.0 ± 1.0 
13.6 ± 0.5 
10.5 ± 0.6 


Average
15.3 ± 1.5 
12.0 ± 1.2 
10.2 ± 1.8 
10.6 ± 1.0 
8.8 ± 0.9


Formula Ia-TP







concentration







(pmol/million cells)b







P valuec
NA
0.2724
0.0507
0.0753
0.0112






aValues are mean ± SEM of duplicate samples collected at each timepoint from two separate experiments.




bAverage Formula Ia -concentration was determined using GraphPad Prism 8.1.2 from the total area under the curve from Formula Ia-TP concentrations detected by LC-MS at 0, 2, 8, 24, and 48 h; and divided by the total time of the assay (48 h).




cP value is determined using one-way ANOVA with Dunnett's multiple comparison analysis of Formula Ia + DMSO compared to various concentrations of CQ or HCQ using GraphPad Prism 8.1.2.














TABLE 4







Effect of CQ or HCQ on RDV-TP Formation in HEp-2 cells









Mean RDV-TP (pmol/million cells)a














1 μM
1 μM
1 μM
1 μM Formula




Formula Ia
Formula Ia
Formula Ia
Ia


Treatment
1 μM
+
+
+
+


time (h)
Formula Ia
1 μM CQ
10 μM CQ
1 μM HCQ
10 μM HCQ





 2
BLQ
BLQ
BLQ
BLQ
BLQ


 8
22.8 ± 1.4
7.7 ± 0.3
3.0 ± 0.5
5.8 ± 1.2
0.4 ± 0.3


24
40.8 ± 5.6
15.9 ± 2.0 
6.2 ± 1.0
15.0 ± 3.2 
3.9 ± 0.8


48
45.9 ± 5.6
20.7 ± 3.2 
10.2 ± 2.0 
20.0 ± 2.5 
7.8 ± 0.6


Average
33.7 ± 4.4
13.6 ± 2.0 
5.8 ± 1.2
12.6 ± 2.3 
3.7 ± 0.6


Formula Ia-TP







concentration







(pmol/million cells)b







P valuec
NA
0.0002
<0.0001
<0.0001
<0.0001






aValues are mean ± SEM of duplicate samples collected at each timepoint from two separate experiments.




bAverage Formula Ia-TP concentration was determined using GraphPad Prism 8.1.2 from the total area under the curve from Formula Ia-TP concentrations detected by LC-MS at 0, 2, 8, 24, and 48 h; and divided by the total time of the assay (48 h).




cP value is determined using one-way ANOVA with Dunnett's multiple comparison analysis of Formula Ia + DMSO compared to various concentrations of CQ or HCQ using GraphPad Prism 8.1.2.







Example 3. SARS-CoV-2 Antiviral Data

The anti-SARS-CoV-2 activities of the compound of Formula Ia and either CQ or HCQ were evaluated in A549-hACE2 transformed airway epithelial cells. The compound of Formula Ia, CQ, and HCQ individually exhibit potent in vitro antiviral activities against SARS-CoV-2, with EC50 values of approximately 59.5 nM, 451 nM, and 365 nM, respectively (Table 5). The compound of Formula Ia EC50 values were not observably different when combined with CQ or HCQ at concentrations up to 2.5 μM (Table 5); however, the overall Nluc signal in the assay decreased in the presence of CQ or HCQ in a dose-dependent manner (FIG. 4A and FIG. 4B). As shown in FIG. 4A, the Nluc signal at 0 μM Formula Ia (DMSO)+CQ corresponds to the Nluc signal at the specific CQ concentration in that treatment condition. The baseline Nluc signal of each Formula Ia titration curve is lowered to levels corresponding to the CQ concentration in that treatment condition. Similar effects are observed in the HCQ-treatment conditions FIG. 4B). At 10 μM of either CQ or HCQ alone, the Nluc signal was fully suppressed, indicating a complete inhibition of SARS-CoV-2 replication by either CQ or HCQ alone. Due to the potent antiviral activity of CQ and HCQ at the 10 μM concentration, a standard EC50 value comparison is unable to reveal the potential decrease in Formula Ia activity against SARS-CoV-2 when combined with CQ or HCQ. Cytotoxicity was not observed at Formula Ia and HCQ combination concentrations analyzed (data not shown).









TABLE 5







SARS-CoV-2 Nluc EC50 values of RDV +


CQ or HCQ in A549-hACE2 cells










Treatment
EC50 (nM)b














Formula Ia +DMSO
59.5



CQ +DMSO
451



HCQ +DMSO
365



Formula Ia + 10 nm CQ
52.0



Formula Ia + 40 nm CQ
52.4



Formula Ia + 160 nm CQ
52.2



Formula Ia + 640 nm CQ
46.0



Formula Ia + 2500 nm CQ
39.8



Formula Ia + 10000 nm CQ
NDc



Formula Ia + 10 nm HCQ
63.6



Formula Ia + 40 nm HCQ
50.7



Formula Ia + 160 nm HCQ
60.0



Formula Ia + 640 nm HCQ
56.2



Formula Ia + 2500 nm HCQ
63.0



Formula Ia + 10000 nm HCQ
NDc








an = 1





bEC50 values were defined in GraphPad Prism 8.1.2 as the concentration at which there was a 50% decrease in the Nluc counts per second (CPS) relative to DMSO vehicle alone (0% virus inhibition) and uninfected control culture (100% virus inhibition).





cND denotes where EC50 values could not be calculated due to complete inhibition of Nluc signal by CQ or HCQ.







Although the foregoing disclosure has been described in some detail by way of illustration and Example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference. Where a conflict exists between the instant application and a reference provided herein, the instant application shall dominate.

Claims
  • 1. A method of treating a viral infection in a human in need thereof, the method comprising administering to the human a therapeutically effective amount of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof:
  • 2. The method of claim 1, wherein the human has not been administered chloroquine, or an analog or salt thereof, within 1 day of receiving a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof.
  • 3. The method of claim 2, wherein the human has not been administered chloroquine, or an analog or salt thereof, within 10 days of receiving the first dose of the compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof.
  • 4. The method of claim 1, further comprising instructing the human not to take chloroquine, or an analog or salt thereof, during the treatment of the viral infection.
  • 5. The method of claim 1, wherein the human is not administered chloroquine, or an analog or salt thereof, during the treatment of the viral infection.
  • 6. The method of claim 1, wherein the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 50 ng/mL, at the time a first dose of the compound of Formula I, Formula Ia, or Formula Ib, or pharmaceutically acceptable salt thereof, is administered to the human.
  • 7. The method of claim 1, wherein the chloroquine, or an analog or salt thereof, is chloroquine, desethylchloroquine, hydroxychloroquine, desethylhydroxychloroquine, or bisdesethylhydroxychloroquine, or a pharmaceutically acceptable salt thereof.
  • 8. The method of claim 7, wherein the chloroquine, or an analog or salt thereof, is chloroquine, or a pharmaceutically acceptable salt thereof.
  • 9. The method of claim 7, wherein the chloroquine, or an analog or salt thereof, is hydroxychloroquine, or a pharmaceutically acceptable salt thereof.
  • 10. The method of claim 1, comprising administering to the human the therapeutically effective amount of the compound of Formula I, or a pharmaceutically acceptable salt thereof:
  • 11. The method of claim 1, wherein the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously.
  • 12. The method of claim 1, wherein the human weighs at least 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 200 mg on day 1, and administered intravenously in a second dose of 100 mg on each of the following 4 days.
  • 13. The method of claim 12, wherein the second dose of 100 mg is administered for an additional 1 to 5 days.
  • 14. The method of claim 1, wherein the human weighs from 3.5 kg to less than 40 kg, and the compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, is administered intravenously in the first dose of 5 mg/kg on day 1, and administered intravenously in a second dose of 2.5 mg/kg on each of the following 4 days.
  • 15. The method of claim 1, wherein the viral infection is caused by an Arenaviridae virus.
  • 16. The method of claim 1, wherein the viral infection is caused by a Coronaviridae virus.
  • 17. The method of claim 16, wherein the Coronaviridae virus is SARS virus, SARS-CoV-2 virus, MERS virus, 229E virus, NL63 virus, OC43 virus, or HKU1 virus.
  • 18. The method of claim 16, wherein the Coronaviridae virus is SARS-CoV-2.
  • 19. The method of claim 16, wherein the viral infection is caused by a virus having at least 70% sequence homology to a viral polymerase selected from the group consisting of SARS-CoV polymerase, SARS-CoV-2 polymerase, and MERS polymerase.
  • 20. The method of claim 1, wherein the viral infection is caused by a Filoviridae virus.
  • 21. The method of claim 20, wherein the Filoviridae virus is Ebola or Marburg.
  • 22. The method of claim 1, wherein the viral infection is caused by a Flaviviridae virus.
  • 23. The method of claim 1, wherein the viral infection is caused by a Pneumoviridae virus.
  • 24. The method of claim 23, wherein the Pneumoviridae virus is respiratory syncytial virus or human metapneumovirus.
  • 25. The method of claim 1, wherein the viral infection is caused by a Paramyxoviridae virus.
  • 26. The method of claim 25, wherein the Paramyxoviridae virus is Nipah or parainfluenza virus.
  • 27. A method of optimizing concentration of a compound of Formula II, or a pharmaceutically acceptable salt thereof, in plasma or blood of a human:
  • 28. The method of claim 27, wherein the human has not been administered chloroquine, or an analog thereof, within 1 day prior to the treatment of the viral infection.
  • 29. The method of claim 27, wherein the human has not been administered chloroquine, or an analog thereof, within 10 days prior to the treatment of the viral infection.
  • 30. The method of claim 27, wherein the human has a plasma or blood concentration of the chloroquine, or an analog or salt thereof, of less than 50 ng/mL, at the time a first dose of the antiviral compound is administered to the human.
  • 31. The method of claim 27, wherein the plasma or blood concentration of the compound of Formula II in the human is higher than a second concentration of a compound of Formula II in a reference human treated with chloroquine, or an analog or salt thereof.
  • 32. The method of claim 27, wherein the antiviral compound is a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof,
  • 33. A method of determining a delivery dose of a compound of Formula I, Formula Ia, or Formula Ib, or a pharmaceutically acceptable salt thereof, for treating a viral infection in a human in need thereof,
  • 34. A method of forming an optimized amount of a compound of Formula II in a human in need thereof, comprising administering to the human a therapeutically effective amount of a compound of Formula Ia, and instructing the human not to take chloroquine, or an analog or salt thereof, wherein upon administration to the human, the compound of Formula Ia is metabolized to the compound of Formula II, wherein the compound of Formula II has the structure:
  • 35. A method of reducing the risk of decreased efficacy of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection
  • 36. A method of preventing a contraindication in a human suffering from a viral infection, the method comprising: (a) determining if the human has taken chloroquine, or an analog or salt thereof, prior to administration of the compound of Formula Ia, or a pharmaceutically acceptable salt thereof,
  • 37. A method of maintaining optimized efficacy of a compound of Formula Ia, or a pharmaceutically acceptable salt thereof, in a human suffering from a viral infection,
  • 38. A method of reducing the risk of a reduced plasma concentration of a compound of Formula II, in a human suffering from a viral infection,
CROSS-REFERENCES TO RELATED APPLICATIONS

This application claims priority to the U.S. Provisional Patent Application No. 63/032,321, filed May 29, 2021, which is incorporated herein in its entirety for all purposes.

US Referenced Citations (180)
Number Name Date Kind
4816570 Farquhar Mar 1989 A
4968788 Farquhar Nov 1990 A
5663159 Starrett, Jr. et al. Sep 1997 A
5792756 Starrett, Jr. et al. Aug 1998 A
6312662 Erion et al. Nov 2001 B1
6475985 Wagner et al. Nov 2002 B1
6476030 Carling et al. Nov 2002 B1
6639059 Kochkine et al. Oct 2003 B1
6656915 Bantia et al. Dec 2003 B1
6909011 Skranc et al. Jun 2005 B2
7078403 Wu et al. Jul 2006 B1
7105493 Sommadossi et al. Sep 2006 B2
7125855 Bhat et al. Oct 2006 B2
7166604 Watson et al. Jan 2007 B2
7176203 Chambers et al. Feb 2007 B2
7268119 Cook et al. Sep 2007 B2
7285658 Cook et al. Oct 2007 B2
7368437 Bojack et al. May 2008 B1
7390791 Becker et al. Jun 2008 B2
7429571 Chand et al. Sep 2008 B2
7514410 Babu et al. Apr 2009 B2
7560434 Babu et al. Jul 2009 B2
7598230 Cook et al. Oct 2009 B2
7608597 Sommadossi et al. Oct 2009 B2
7713941 Cook et al. May 2010 B2
7803788 Becker et al. Sep 2010 B2
7807653 Cook et al. Oct 2010 B2
7842672 Boojamra et al. Nov 2010 B2
7951787 McGuigan May 2011 B2
7973013 Cho et al. Jul 2011 B2
7994139 Babu et al. Aug 2011 B2
8008264 Butler et al. Aug 2011 B2
8012941 Cho et al. Sep 2011 B2
8012942 Butler et al. Sep 2011 B2
8071568 Narjes et al. Dec 2011 B2
8119607 Francom et al. Feb 2012 B2
8242085 Babu et al. Aug 2012 B2
8318682 Butler et al. Nov 2012 B2
8415308 Cho et al. Apr 2013 B2
8455451 Cho et al. Jun 2013 B2
8853171 Butler et al. Oct 2014 B2
8871737 Smith et al. Oct 2014 B2
8889159 Clearly et al. Nov 2014 B2
8980865 Wang Mar 2015 B2
9090642 Cho et al. Jul 2015 B2
9243022 Beigelman et al. Jan 2016 B2
9249174 Beigelman et al. Feb 2016 B2
9278990 Smith et al. Mar 2016 B2
9388208 Clarke et al. Jul 2016 B2
9393256 Ray et al. Jul 2016 B2
9452154 Delaney et al. Sep 2016 B2
9481703 Kalayanov et al. Nov 2016 B2
9487544 Cho et al. Nov 2016 B2
9504701 Casola et al. Nov 2016 B2
9540411 Kalayanov et al. Jan 2017 B2
9549941 Cleary et al. Jan 2017 B2
9605018 Wang et al. Mar 2017 B2
9616076 Casola et al. Apr 2017 B2
9701682 Clarke et al. Jul 2017 B2
9724360 Chun et al. Aug 2017 B2
9828408 Kalayanov Nov 2017 B2
RE46762 Butler et al. Mar 2018 E
9938283 Pandey et al. Apr 2018 B2
9949994 Chun et al. Apr 2018 B2
10023600 Butler et al. Jul 2018 B2
10034893 Luly et al. Jul 2018 B2
10059716 Clarke et al. Aug 2018 B2
10065958 Mackman et al. Sep 2018 B2
10251898 Chun et al. Apr 2019 B2
10251904 Clarke et al. Apr 2019 B2
10377761 Clarke et al. Aug 2019 B2
RE47589 McGuigan Sep 2019 E
10675296 Larson Jun 2020 B2
10682368 Perron et al. Jun 2020 B2
10695357 Chun et al. Jun 2020 B2
10695361 Clarke et al. Jun 2020 B2
10696679 Mackman et al. Jun 2020 B2
10836787 Brak et al. Nov 2020 B2
10988498 Butler et al. Apr 2021 B2
11007208 Clarke et al. May 2021 B2
11260070 Perron et al. Mar 2022 B2
11266666 Chun et al. Mar 2022 B2
11266681 Larson et al. Mar 2022 B2
11344565 Axt et al. May 2022 B2
11377456 Souza et al. Jul 2022 B2
11382926 Clarke et al. Jul 2022 B2
11492353 Mackman et al. Nov 2022 B2
11541071 Liang et al. Jan 2023 B1
11597742 Brak et al. Mar 2023 B2
11613553 Badalov et al. Mar 2023 B2
11660307 Cihlar et al. May 2023 B2
11814406 Bunyan et al. Nov 2023 B2
11845755 Bartlett et al. Dec 2023 B2
11851438 Bartlett et al. Dec 2023 B2
20030050229 Sommadossi et al. Mar 2003 A1
20030092775 Ernst et al. May 2003 A1
20040006002 Sommadossi et al. Jan 2004 A1
20040023901 Cook et al. Feb 2004 A1
20040063658 Roberts et al. Apr 2004 A1
20040067901 Bhat et al. Apr 2004 A1
20040138170 Montgomery et al. Jul 2004 A1
20050187180 Loeb et al. Aug 2005 A1
20050209166 Eckhardt et al. Sep 2005 A1
20050215513 Boojamra et al. Sep 2005 A1
20050250728 Bantia et al. Nov 2005 A1
20060058303 Chambers et al. Mar 2006 A1
20060142238 McGuigan Jun 2006 A1
20060241064 Roberts et al. Oct 2006 A1
20080107628 Boojamra et al. May 2008 A1
20080161324 Johansen et al. Jul 2008 A1
20080280842 MacCoss et al. Nov 2008 A1
20090004138 Francom et al. Jan 2009 A1
20090221524 Kotra et al. Sep 2009 A1
20090233879 Reddy et al. Sep 2009 A1
20090317361 Cho et al. Dec 2009 A1
20100015094 Babu et al. Jan 2010 A1
20100016251 Sofia et al. Jan 2010 A1
20100021425 Butler et al. Jan 2010 A1
20100035835 Narjes et al. Feb 2010 A1
20100035836 Francom et al. Feb 2010 A1
20100065512 Bjorsvik Mar 2010 A1
20100129437 Gaillard May 2010 A1
20100203015 Butler et al. Aug 2010 A1
20100234584 Chang Sep 2010 A1
20100249068 Beigelman et al. Sep 2010 A1
20100291031 Francom et al. Nov 2010 A2
20100298257 Ross et al. Nov 2010 A1
20100305202 Hwang et al. Dec 2010 A1
20110070194 Cho et al. Mar 2011 A1
20110084230 Knochel et al. Apr 2011 A1
20110230654 Butler et al. Sep 2011 A1
20110257122 Sofia et al. Oct 2011 A1
20110293563 Butler et al. Dec 2011 A1
20120009147 Cho et al. Jan 2012 A1
20120020921 Cho et al. Jan 2012 A1
20120027752 Mackman et al. Feb 2012 A1
20120071434 Smith et al. Mar 2012 A1
20120107274 Clarke et al. May 2012 A1
20130034521 Butler et al. Feb 2013 A1
20130143835 Eneroth et al. Jun 2013 A1
20130281686 Cho et al. Oct 2013 A1
20130315868 Mayes Nov 2013 A1
20130344028 Butler et al. Dec 2013 A2
20140219958 Luly et al. Aug 2014 A1
20150031687 Guo et al. Jan 2015 A1
20150111839 Mackman et al. Apr 2015 A1
20150133395 Clarke et al. May 2015 A1
20150152116 Mackman et al. Jun 2015 A1
20150210682 Han et al. Jul 2015 A1
20150252057 Guo et al. Sep 2015 A1
20160058779 Casola et al. Mar 2016 A1
20160122344 Han et al. May 2016 A1
20160122356 Axt et al. May 2016 A1
20160122374 Chun May 2016 A1
20160176899 Schwitter et al. Jun 2016 A1
20160220586 Andre et al. Aug 2016 A1
20160237090 Hu et al. Aug 2016 A1
20170071964 Clark et al. Mar 2017 A1
20180346504 Brak et al. Dec 2018 A1
20190083525 Larson Mar 2019 A1
20200197422 Axt et al. Jun 2020 A1
20200360420 Larson Nov 2020 A1
20200376014 Perron et al. Dec 2020 A1
20210052613 Chun et al. Feb 2021 A1
20210061806 Mackman et al. Mar 2021 A1
20210283150 Cihlar et al. Sep 2021 A1
20210309689 Badalov et al. Oct 2021 A1
20210330685 Ellis et al. Oct 2021 A1
20210393659 O'Neil et al. Dec 2021 A1
20210403497 Butler et al. Dec 2021 A1
20220081462 Chun et al. Mar 2022 A1
20220280549 Larson et al. Sep 2022 A1
20220354873 Axt et al. Nov 2022 A1
20220356196 Byun et al. Nov 2022 A1
20230027727 Clarke et al. Jan 2023 A1
20230040586 Byun et al. Feb 2023 A1
20230125751 Mackman et al. Apr 2023 A1
20230151043 Bunyan et al. May 2023 A1
20240009220 Bannister et al. Jan 2024 A1
20240024341 Ellis et al. Jan 2024 A1
Foreign Referenced Citations (170)
Number Date Country
1080841952 May 2018 CN
WO2018099946 Jun 2018 CN
110330540 Oct 2019 CN
110724174 Jan 2020 CN
110776512 Feb 2020 CN
111171078 May 2020 CN
111205294 May 2020 CN
111205327 May 2020 CN
111233869 Jun 2020 CN
111265532 Jun 2020 CN
111440176 Jul 2020 CN
111548384 Aug 2020 CN
111961057 Nov 2020 CN
202011613943.3 Dec 2020 CN
112778310 May 2021 CN
202110562244.9 May 2021 CN
113754665 Jun 2021 CN
113185519 Jul 2021 CN
113248508 Aug 2021 CN
113292565 Aug 2021 CN
113387954 Sep 2021 CN
113735862 Sep 2021 CN
114292272 Dec 2021 CN
114437159 May 2022 CN
114621229 Jun 2022 CN
114765979 Jul 2022 CN
202121023147 May 2021 IN
202134041493 Sep 2021 IN
202011021676 Nov 2021 IN
2005185235 Jul 2005 JP
2005187428 Jul 2005 JP
WO1991019721 Dec 1991 WO
WO1999045029 Sep 1999 WO
WO2000056734 Sep 2000 WO
WO200075157 Dec 2000 WO
WO2001032153 May 2001 WO
WO2001060315 Aug 2001 WO
WO2001090121 Nov 2001 WO
WO2001091737 Dec 2001 WO
WO2001092282 Dec 2001 WO
WO2002008241 Jan 2002 WO
WO2002018404 Mar 2002 WO
WO2002032920 Apr 2002 WO
WO2002057287 Jul 2002 WO
WO2002057425 Jul 2002 WO
WO2003093272 Nov 2003 WO
WO2003093273 Nov 2003 WO
WO2003100009 Dec 2003 WO
WO2004046159 Jun 2004 WO
WO2004046331 Jun 2004 WO
WO2004112687 Dec 2004 WO
WO2005009418 Feb 2005 WO
WO2005092877 Oct 2005 WO
WO2005123087 Dec 2005 WO
WO2006031725 Mar 2006 WO
WO2006050161 May 2006 WO
WO2006064033 Jun 2006 WO
WO2006065335 Jun 2006 WO
WO2006121820 Nov 2006 WO
WO2006135978 Dec 2006 WO
WO2007027248 Mar 2007 WO
WO2007056170 May 2007 WO
WO2007062542 Jun 2007 WO
WO2007064883 Jun 2007 WO
WO2007064931 Jun 2007 WO
WO2007065289 Jun 2007 WO
WO2007065829 Jun 2007 WO
WO2007095269 Aug 2007 WO
WO2007097991 Aug 2007 WO
WO2007113294 Oct 2007 WO
WO2007135134 Nov 2007 WO
WO2008005542 Jan 2008 WO
WO2008011406 Jan 2008 WO
WO2008055870 May 2008 WO
WO2008079206 Jul 2008 WO
WO2008082601 Jul 2008 WO
WO2008085508 Jul 2008 WO
WO2008089105 Jul 2008 WO
WO2008116064 Sep 2008 WO
WO2008121634 Oct 2008 WO
WO2008141079 Nov 2008 WO
WO2009009951 Jan 2009 WO
WO2009018609 Feb 2009 WO
WO2009131926 Oct 2009 WO
WO2009132123 Oct 2009 WO
WO2009132135 Oct 2009 WO
WO2010002877 Jan 2010 WO
WO2010036407 Apr 2010 WO
WO2010039548 Apr 2010 WO
WO2010093608 Aug 2010 WO
WO2010099458 Sep 2010 WO
WO2010108140 Sep 2010 WO
WO2010135569 Nov 2010 WO
WO2011011303 Jan 2011 WO
WO2010111381 Mar 2011 WO
WO2011035231 Mar 2011 WO
WO2011035250 Mar 2011 WO
WO2011080568 Jul 2011 WO
WO2011100131 Aug 2011 WO
WO2011123645 Oct 2011 WO
WO2011123668 Oct 2011 WO
WO2011123672 Oct 2011 WO
WO2011150288 Dec 2011 WO
WO2012012465 Jan 2012 WO
WO2012012776 Jan 2012 WO
WO2012039787 Mar 2012 WO
WO2012039791 Mar 2012 WO
WO2012051570 Apr 2012 WO
WO2012040127 May 2012 WO
WO2012121764 Sep 2012 WO
WO2012142523 Oct 2012 WO
WO2012158643 Nov 2012 WO
WO2013039861 Mar 2013 WO
WO2013084165 Jun 2013 WO
WO2014033617 Mar 2014 WO
WO2014042433 Mar 2014 WO
WO2014078463 May 2014 WO
WO2014078778 May 2014 WO
WO2014116755 Jul 2014 WO
WO2014169280 Oct 2014 WO
WO2014209979 Dec 2014 WO
WO2016107833 Dec 2014 WO
WO2015054465 Apr 2015 WO
WO2015069939 May 2015 WO
WO2015173164 Nov 2015 WO
WO2015200205 Dec 2015 WO
WO2015200219 Dec 2015 WO
WO2016012470 Jan 2016 WO
WO2016023877 Feb 2016 WO
WO2016069825 May 2016 WO
WO2016069826 May 2016 WO
WO2016069827 May 2016 WO
WO2016102438 Jun 2016 WO
WO2016107832 Jul 2016 WO
WO2016120186 Aug 2016 WO
WO2016128335 Aug 2016 WO
WO2017165489 Sep 2017 WO
WO2017184668 Oct 2017 WO
WO2018085307 May 2018 WO
WO2018121678 Jul 2018 WO
WO2018145148 Aug 2018 WO
WO2018204198 Nov 2018 WO
WO2019014247 Jan 2019 WO
WO2019053696 Mar 2019 WO
WO2022098371 Nov 2020 WO
WO2021021717 Feb 2021 WO
WO2021040356 Mar 2021 WO
WO2021050961 Mar 2021 WO
WO2021147236 Jul 2021 WO
WO2021175296 Sep 2021 WO
WO2021195661 Sep 2021 WO
WO2022142477 Sep 2021 WO
WO2021202907 Oct 2021 WO
WO2021207049 Oct 2021 WO
WO2021213288 Oct 2021 WO
WO2021222807 Nov 2021 WO
WO2022143473 Dec 2021 WO
WO2022008642 Jan 2022 WO
WO2022029704 Feb 2022 WO
WO2022047065 Mar 2022 WO
WO2022047441 Mar 2022 WO
WO2022165386 Aug 2022 WO
WO2022174194 Aug 2022 WO
WO2022217153 Oct 2022 WO
WO2022217154 Oct 2022 WO
WO2022217155 Oct 2022 WO
WO2022218274 Oct 2022 WO
WO2022222994 Oct 2022 WO
WO2022251663 Dec 2022 WO
WO2022265964 Dec 2022 WO
Non-Patent Literature Citations (447)
Entry
Anonymous [online], “University of Alabama & Multi-Center Collaboration Help Develop Remdesivir with Gilead Thanks to $37.5m from NIH.” TrialSiteNews.com, retrieved on Mar. 13, 2023, URL <https://www.trialsitenews.com/a/university-of-alabama-multi-center-collaboration-help-develop-remdesivir-with-gilead-thanks-to-37-5m-from-nih>, Mar. 1, 2020, 5 pages.
Jonckers et al., “2′Deoxy-2′-spirocyclopropylcytidine Revisited: A New and Selective Inhibitor of the Hepatitis C Virus NS5B Polymerase,” Journal of Medicinal Chemistry, Nov. 2010, 53(22)8150-60.
Jones et al., “Di- and Triester Prodrugs of the Varicella-Zoster Antiviral Agent 6-Methoxypurine Arabinoside,” Journal of Medicinal Chemistry, Jan. 1992, 35(1):56-63.
Joseph [online], “As the coronavirus spreads, a drug that once raised the world's hopes is given a second shot,” StatNews.com, retrieved on Mar. 13, 2023, URL <https:/www.statnews.com/2020/03/16/remdesivir-surges-ahead-against-coronavirus>, Mar. 16, 2020. 11 pages.
Kim et al., “Synthesis and Evaluation of 2-Amino-6-fluoro-9-(4-hydroxy-3-hydroxymethylbut-1-y1) purine Mono-and Diesters as Potential Prodrugs of Penciclovir,” Bioorganic & Medicinal Chemistry, Mar. 1999, 7(3):565-70.
Kim et al., “Synthesis and Evaluation of 2-Amino-9-(1,3-dihydroxy-2-propoxymethyl)-6-fluoropurine Mono-and Diesters as Potential Prodrugs of Ganciclovir,” Journal of Medicinal Chemistry, Jan. 1999, 42(2):324-28.
Klumpp et al., “Chapter 20: Discovery and Clinical Evaluation of the Nucleoside Analog Balapiravir (R1626) for the Treatment of HCV Infection,” Antiviral Drugs: From Basic Discovery through Clinical Trials, Jun. 20, 2011, pp. 287-304.
Koplon [online], “$37.5 million grant will address research of high-priority infections,” UAB News, retrieve on Mar. 13, 2023, URL <https://www.uab.edu/news/health/item/10307-37-5-million-grant-will-address-research-of-high-priority-infections>, Mar. 20, 2019, 1 page.
Moorman et al., “5′-ester prodrugs of the varicella-zoster antiviral agent, 6-methoxypurine arabinoside,” Antiviral Chemistry & Chemotherapy, Jun. 1992, 3(3):141-46.
Nilsson et al., “Discovery of 4′-azido-2′-deoxy-2′-C-methyl cytidine and prodrugs thereof: A potent inhibitor of Hepatitis C virus replication,” Bioorganic & Medicinal Chemistry Letters, May 2012, 22(9):3265-68.
Remington's Pharmaceutical Science, 17th ed., Gennaro (ed)., 1985, Chapter 68, 58 pages.
Xu et al., “Off-Target In Vitro Profiling Demonstrates that Remdesivir Is a Highly Selective Antiviral Agent,” Antimicrobial Agents and Chemotherapy, Jan. 20, 2021, 65(2), 14 pages.
Yan et al., “Advantages of the Parent Nucleoside GS-441524 over Remdesivir for Covid-19 Treatment,” ACS Medicinal Chemistry Letters, Jun. 30, 2020, 11(7):1361-1366.
Yan et al., “Gilead should ditch remdesivir and focus on its simpler and safer ancestor,” STAT Health Care News, May 14, 2020, 6 pages.
Yan et al., “Pharmacokinetics of 1 Orally Administered GS-441524 in Dogs,” bioRxiv Preprint, May 31, 2021, 18 pages.
U.S. Appl. No. 17/665,724, filed Feb. 7, 2022, Steven Donald Axt.
U.S. Appl. No. 14/746,430, filed Jun. 22, 2015, Aesop Cho.
U.S. Appl. No. 17/458,023, filed Aug. 26, 2021, Byoung-Kwon Chun.
U.S. Appl. No. 18/098,950, filed Jan. 19, 2023, Byoung-Kwon Chun.
Beer et al., “Characteristics of Filoviridae: Marburg and Ebola Viruses,” Naturwissenschaften, 1999, 86:8-17.
Brands et al., “Crystallization-Induced Diastereomer Transformations,” Chem. Rev., 2006, 106(7):2711-2733.
Brotschi et al., “Bipyridyl and biphenyl DNA: A recognition motif based on interstrand aromatic stacking,” Chemistry—A European Journal, 2005, 11(6):1911-1923.
Carey et al., “Addition, Condensation and Substitution Reactions of Carbonyl Compounds,” Advanced Organic Chemistry: Part B: Reaction and Synthesis, Springer Science & Business Media, 2007, pp. 629-711.
Gordon et al., “Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency,” J. Biol. Chem., 2020, 295(20):6785-6797.
Gordon et al., “The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus,” Journal of Biol. Chemistry, 2020, 295(15):4773- 4779.
Leyssen et al., “Molecular strategies to inhibit the replication of RNA Viruses,” Antiviral Research, 2008, 78:9-25.
McGuigan et al., “Design, synthesis and biological evaluation of phosphorodiamidate prodrugs of antiviral and anticancer nucleosides,” European Journal of Medical Chemistry, 2013, 70:326-340.
Pruijssers et al., “Remdesivir Inhibits SARS-CoV-2 in Human Lung Cells and Chimeric SARS-CoV Expressing the SARS-CoV-2 RNA Polymerase in Mice,” Cell Reports, 2020, 32(107940):1-16.
Sheahan et al., “Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MER-CoV,” Nature Communications, 2020, 11(222):1-14.
Tschesnokov et al., “Template-dependent inhibition of coronavirus RNA-dependent RNA polymerase by remdesivir reveals a second mechanism of action,” J. Biol. Chem., 2020, 295(47):16156-16165.
Yang et al., “Lewis acid catalyzed direct cyanation of indoles and pyrroles with N-cyano-N-phenyl-p-toluenesulfonamide (NCTS),” Organic Letters, 2011, 13(20): 5608-5611.
fda.gov [online], “Remdesivir by Gilead Sciences: FDA Warns of Newly Discovered Potential Drug Interaction That May Reduce Effectiveness of Treatment,” Jun. 15, 2020, retrieved on Sep. 2, 2022, retrieved from URL <https://www.fda.gov/safety/medical-product-safety-information/remdesivir-gilead-sciences-fda-warns-newly-discovered-potential-drug-interaction-may-reduce>, 2 pages.
Khan et al., “Coronaviruses disease 2019 (COVID-19): Causative agent, mental health concerns, and potential management options,” Journal of Infection and Public Health, Dec. 2020, 13(12):1840-1844.
Kulli, “K Banhatti Polynomials of Remdesivir, Chloroquine, Hydroxychloroquine: Research Advances for the Prevention and Treatment of COVID-19,” SSRG International Journal of Applied Chemistry, May-Aug. 2020, 7(2):48-55.
Liu et al., “Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro.” Cell Discovery, Mar. 18, 2020, 6:16, 4 pages.
Owusu et al., “A Comparison Analysis on Remdesivir, Favipiravir, Hydroxychloroquine, Chloroquine and Azithromycin in the Treatment of Corona Virus Disease 2019 (COVID-19)—A Review,” World Journal of Pharmacy and Pharmaceutical Sciences, May 2020, 9(5):121-133.
Pizzorno et al., “In vitro evaluation of antiviral activity of single and combined repurposable drugs against SARS-CoV-2.” Antiviral Research, Sep. 2020, 181:104878.
Rebeaud et al., “SARS-CoV-2 and the Use of Chloroquine as an Antiviral Treatment,” Frontiers in Medicine, Apr. 24, 2020, 7:184, 6 pages.
Taiwanese Office Action in TW Appln. No. 110119391, dated Aug. 22, 2022, 17 pages (with English translation).
Vieira et al., “Development of a Large-Scale Cyanation Process Using Continuous Flow Chemistry En Route to the Synthesis of Remdesivir,” Organic Process Research & Development, May 2020, 24(10):2113-2121.
Wang et al., “Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial,” Lancet, Apr. 29, 2020, 395:1569-1578.
Agostini et al., “Coronavirus Susceptibility to the Antiviral Remdesivir (GS5734) Is Mediated by the Viral Polymerase and the Proofreading Exoribonuclease”, MBIO, Mar. 6, 2018, 9(2):1-15.
Alessandrini, et al., Synthesis of Differently Protected 1-C-methyl-ribofuranoses Intermediates for the Preparation of Biologically Active 1′-C-methyl-ribonucleosides, Journal of Carbohydrate Chemistry, 2008, pp. 332-344, vol. 27, No. 5.
Ali, et al., Quantitative structure-activity relationships (QSAR) of two series of O-aryl or N-acyl O-ethyl phosphoramidate and phosphorodiamidate fungicides incorporating amino acid ethyl esters, Bulletin of Environmental Contamination and Toxicology, 2000, pp. 415-420, vol. 65, No. 4.
Anonymous, “Gillings research on broad-spectrum antiviral could aid public health response to coronavirus outbreaks”, -UNC Gillings School of Global Public Health, Jan. 10, 2020, retrieved on May 13, 2021, revrieved from URL <“https://sph.unc.edu/sph-news/gillings-research-on-broad-spectrum-antiviral-could-aid-public-health-response-to-coronavirus-outbreaks/”>, 5 pages.
Arimilli, et al., Synthesis, In Vitro Biological Evaluation and Oral Bioavailability of 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) Prodrugs, Antiviral Chemistry & Chemotherapy, 1997, pp. 557-564, vol. 8, No. 6.
Asbun, et al., Synthesis of 5-substituted Pyrimidines. II, Journal of Organic Chemistry, 1968, pp. 140-142, vol. 31.
Ballini, et al., Enantioselective Synthesis of the Lactone Moiety of the Mevinic Acids using D-Xylose as a Chiral Precursor, Journal of the Chemical Society, Perkin Transactions 1, 1991, pp. 490-491.
Balzarini, et al., Inhibition of Feline (FIPV) and Human (SARS) Coronavirus by Semisynthetic Derivatives of Glycopeptide Antibiotics, Antiviral Research, 2006, pp. 20-33, vol. 72.
Bandini, et al., Indium tribromide: a highly effective catalyst for the addition of trimethylsilyl cyanide to α-hetero-substituted ketone, Tetrahedron Letters, 2001, pp. 3041-3043. vol. 42.
Barker, et al., 2,3,5-Tri-O-benzyl-D-ribosyl and -L-arabinosyl Bromides, Journal of Organic Chemistry, 1961, pp. 4605-4609, vol. 26, No. 11.
Barl, et al., The halogen/magnesium-exchange using iPrMgCl⋅LiCl and related exchange reagents, Heterocycles, Jan. 2014, pp. 827-844, vol. 88, No. 2.
Belokon, et al., Optimized catalysts for the asymmetric addition of trimethylsilyl cyanide to aldehydes and ketones, Tetrahedron, 2001, pp. 771-779, vol. 57.
Benksim, et al., A Novel Stereospecific Synthesis of Glycosyl Cyanides from 1,2-O-sulfinyl Derivatives, Organic Letters, 2004, pp. 3913-3915, vol. 6, No. 22.
Benzaria, et al., Synthesis, In Vitro Antiviral Evaluation, and Stability Studies of Bis(S-acyl-2-thioethyl) Ester Derivatives of 9-[2-(phosphonomethoxy )ethyl]adenine (PMEA) as Potential PMEA prodrugs with Improved Oral Bioavailability, J. Med. Chem., 1996, pp. 4958-4965, vol. 39, No. 25.
Bio, et al., Practical Synthesis of a Potent Hepatitis C Virus RNA Replication Inhibitor, J. Org. Chem., 2004, pp. 6257-6266, vol. 69, No. 19.
Bobeck, et al., Advances in Nucleoside Monophosphate Prodrugs as Anti-HCV Agents, Antiviral Therapy, 2010, pp. 935-950, vol. 15.
Bobrowski et al., “Synergistic and Antagonistic Drug Combinations against SARS-CoV-2”, Molecular Therapy, Feb. 2021, 29(2):873-885.
Bojack, et al., Design and Synthesis of Inhibitors of Adenosine and AMP Deaminases, Organic Letters, 2001, pp. 839-842, vol. 3, No. 6.
Boyer, et al., Pathogenesis, diagnosis and management of hepatitis C, Journal of Hepatology, 2000, pp. 98-112, vol. 32.
Bozza, Zika Outbreak, Brazil 2015, ISARIC, 2015, 28 pages.
Bradley et al., “The Management of Community-Acquired Pneumonia in Infants and Children Older Than 3 Months of Age: Clinical Practice Guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America”, Pediatric Community Pneumonia Guidelines, Clinical Infectious Diseases, Oct. 2011, 53(7):e25-e76.
Brittain, Polymorphism in Pharmaceutical Solids, 2nd Edition, 2009, pp. 183-226, Informa Healthcare USA, Inc.
Brown et al., “Broad spectrum antiviral remdesivir inhibits human endemic and zoonotic deltacoronaviruses with a highly divergent RNA dependent RNA polymerase”, Antiviral Research, Jun. 21, 2019, 169:1-31.
Brown, Progress towards improving antiviral therapy for hepatitis C virus polymerase inhibitors. Part O: Nucleoside analogues, 2009, pp. 709-725, vol. 18.
Bullard-Feibelman, et al., The FDA-approved drug Sofosbuvir inhibits Zika Virus infection, Antiviral Res., Jan. 1, 2018, pp. 134-140, vol. 137.
Burns, A Glimmer of Hope for Fatal Feline Disease, JAVMAnews, Dec. 15, 2017, 5 pages.
Butora, et al., Synthesis and HCV inhibitory properties of 9-deaza- and 7,9-dideaza-7-oxa-2′-C-methyladenosine, Bioorganic & Medicinal Chemistry, 2007, pp. 5219-5229, vol. 15, No. 15.
Cabirol, et al., Robust and Efficient, yet Uncatalyzed, Synthesis of Triarylsilyl-protected Cyanohydrins from Ketones, 2008, pp. 2446-2449, vol. 73.
Caira, Crystalline Polymorphism of Organic Compounds, Topics in Current Chemistry, 1998, pp. 163-208, vol. 198.
Calès, et al., Treatment of liver fibrosis: clinical aspects, Gastroentérologie Clinique et Biologique, 2009, pp. 958-966, vol. 33, No. 10-11.
Calisher, et al., Antigenic Relationships between Flaviviruses as Determined by Cross-neutralization Tests with Polyclonal Antisera, Journal of General Virology, 1989, pp. 37-43, vol. 70.
Camps, Studies on Structurally Simple -αß-butenolides-II, Tetrahedron, 1982, pp. 2395-2402, vol. 38, No. 15.
Carroll, Robust Antiviral Efficacy upon Administration of a Nucleoside Analog to Hepatitis C Virus-Infected Chimpanzees, Antimicrobial Agents and Chemotherapy, 2009, pp. 926-934, vol. 53, No. 3.
Carryer et al., “The effect of cortisone on bronchial asthma and hay fever occurring in subjects sensitive to ragweed pollen”, Journal of Allergy, Jul. 1950, 21(4): 282-287.
CAS No. 1476-52-4, “Desethyl Chloroquine”, ChemSRc, retrieved on Jul. 29, 2021, retrieved from URL <“https://www.chemsrc.com/en/cas/1476-52-4_1032909.html”>, 5 pages.
CAS No. 4298-15-1, “2-[4-[(7-chloroquinolin-4-yl)amino]pentylamino]ethanol”, ChemSRc, retrieved on Jul. 29, 2021, retrieved from URL <“https://www.chemsrc.com/en/cas/4298-15-1_589766.html”>, 4 pages.
CAS No. 54-05-7, “Chloroquine”, ChemSRc, retrieved on Jul. 29, 2021, retrieved from URL <“https://www.chemsrc.com/en/cas/54-05-7_419322.html”>, 16 pages.
CAS Registry No. 1809249-37-3, “L-Alanine, N-[(S)-hydroxyphenoxyphosphinyl]-, 2-ethylbutyl ester, 6-ester with 2-C-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-2,5-anhydro-D-altrononitrile”, American Cemical Society, retrieved on Jul. 27, 2021, retrieved from URL <“https://commonchemistry.cas.org/detail?cas_rn=1809249-37-3”>, 3 pages.
Chapman, et al., RSV604, a Novel Inhibitor of Respiratory Syncytial Virus Replication, Antimicrobial Agents and Chemotherapy, 2007, pp. 3346-3353, vol. 51, No. 9.
Cho, et al., “Synthesis and antiviral Activity of a Series of 1′-Substituted 4-aza-7,9-dideazaadenosine C-Nucleosides”, Bioorganic & Medicinal Chemistry Letters, 2012, 22:2705-2707.
Cho, et al., Discovery of the First C-Nucleoside HCV Polymerase Inhibitor (GS-6620) with Demonstrated Antiviral Response in HCV Infected Patients, J. Med. Chem., 2014, pp. 1812-1825, vol. 57, No. 5.
Cihlar, et al., Design and Profiling of GS-9148, a Novel Nucleotide Analog Active against Nucleoside-resistant Variants of Human Immunodeficiency Virus Type 1, and Its Orally Bioavailable Phosphonoamidate Prodrug, GS-9131, Antimicrobial Agents and Chemotherapy, 2008, pp. 655-665, vol. 52, No. 2.
Clark, et al., Design, Synthesis, and Antiviral Activity of 2′-Deoxy-2′-fluoro-2′-C-methylcytidine, a Potent Inhibitor of Hepatitis C Virus Replication, Journal of Medicinal Chemistry, 2005, pp. 5504-5508, vol. 48, No. 17.
Clarke, et al., Discovery of beta-D-2′-Deoxy-2′-alpha-Fluoro-4′-alpha-Cyano-5-aza-7,9-Dideaza Adenosine as a Potent Nucleoside Inhibitor of Respiratory Syncytial Virus with Excellent Selectivity Over Mitochondrial RNA and DNA Polymerases, Bioorganic & Medicinal Chemistry Letters, Apr. 29, 2015, pp. 2484-2487, vol. 25.
Colacino, et al., Synthesis and Biological Evaluation of Some 5-Nitro- and 5-Amino Derivatives of 2′-Deoxycytidine, 2′,3′-Dideoxyuridine, and 2′,3′-Dideoxycytidine, Nucleoside, Nucleotides & Nucleic Acids, 2003, pp. 2013-2026, vol. 22, No. 11.
Dai, et al., Synthesis of 2′-C-α-Fluoromethyluridine, Organic Letters, 2003, pp. 807-810, vol. 5, No. 6.
Damont et al., “Synthesis of 1′-C-Fluoromethyladenosine,” Nucleosides, Nucleotides , and Nucleic Acids, 2007, 26:1431-1434.
De Clercq, Antiviral Drugs: Current State of the Art, J. Clin. Virol., 2001, pp. 73-89, vol. 22, No. 1.
De Clercq, Molecular Targets for Antiviral Agents, The Journal of Pharmacology and Experimental Therapeutics, 2001, pp. 1-10, vol. 297, No. 1.
De Francesco, et al., Approaching a New Era for Hepatitis C Virus Therapy: Inhibitors of the NS3-4A Serine Protease and the NS5B RNA-Dependent RNA Polymerase, Antiviral Research, 2003, pp. 1-16, vol. 58, No. 1.
De Las Heras, Synthesis of Ribosyl and Arabinosyl Cyanides by Reaction of 1-O-Acyl Sugars with Trimethylsilyl Cyanide, Journal of the Chemical Society, Perkin Transactions 1, 1982, pp. 903-907.
De Lombaert, et al., N-Phosphonomethyl Dipeptides and Their Phosphonate Prodrugs, a New Generation of Neutral Endopeptidase (NEP, EC 3.4.24.11) Inhibitors, J. Med. Chem., 1994, pp. 498-511, vol. 37, No. 4.
Di Bisceglie, et al., The Unmet Challenges of Hepatitis C, Scientific American, Oct. 1999, pp. 80-85.
Dolzhenko, et al., Pyrazolo[1,5-a][1,3,5]Triazines(5-Aza-9-Deazapurines): Synthesis and Biological Activity, Heterocycles, 2008, pp. 1575-1622, vol. 75, No. 7.
Domingo, et al., The quasispecies (extremely heterogeneous) nature of viral RNA genome populations: biological relevance—a review, Gene, 1985, pp. 1-8, vol. 40.
Dondoni, et al., Thiazole-Based Synthesis of Formyl C-Glycosides, Journal of Organic Chemistry, 1994, pp. 6404-6414, vol. 59.
Dudfield, et al., Synthesis of C-ribosyl 1,2,4-triazolo[3,4-f][1,2,4]triazines as Inhibitors of Adenosine and AMP Deaminasses, J. Chem. Soc., Perkin Trans. 1, 1999, pp. 2937-2942.
Dudfield, P. et al., Synthesis of C-ribosyl imidazo[2,1-f][1,2,4]triazines as inhibitors of adenosine and AMP deaminases, J. Chem. Soc, Perkin Trans I, 1999, pp. 2929-2936.
Durcan et al., “Hydroxychloroquine Blood Levels in Systemic Lupus Erythematosus: Clarifying Dosing Controversies and Improving Adherence”, Journal of Rheumatology, 2015, 42(11):2092-2097.
Dymock, et al., Novel approaches to the treatment of hepatitis C virus infection, Antiviral Chemistry & Chemotherapy, 2000, pp. 79-96, vol. 11, No. 2.
El Safadi, et al., 5-Modified-2′-dU and 2′-dC as Mutagenic Anti HIV-1 Proliferation Agents: Synthesis and Activity, Journal of Medicinal Chemistry, 2010, pp. 1534-1545, vol. 53, No. 4.
Farquhar, et al., “Biologically Reversible Phosphate-Protective Groups”, Journal of Pharmaceutical Sciences, 1983, pp. 324-325, vol. 72, No. 3.
Fauquet et al., “Abbreviations for vertebrate virus species names”, Archives of Virology, Dec. 31, 1999, pp. 1865-1880.
Foster et al., “Deuterium isotope effects in studies of drug metabolism,” Trends in Pharmacological Sciences, Jan. 1984, 5:524-527.
Franchetti et al., Antitumor Activity of C-Methyl-β-D-ribofuranosyladenine Nuceoside Ribonuceotide Reductase Inhibitors, J. Med. Chem. 2005, pp. 4983-4989, vol. 48.
Fukumoto, et al., Viral Dynamics of Hepatitis C Early After Orthotopic Liver Transplantation: Evidence for Rapid Turnover of Serum Virions, Hepatology, 1996, pp. 1351-1354, vol. 24.
Garcia, et al., Synthesis of (2,3,4,6-tetra-O-acetyl-alpha-D-glycopyranosyl)thiophene derivatives as new C-nucleoside analogues, J. Carbohydrate Chemistry, 2001, pp. 681-687, vol. 20, No. 7/8.
Gardelli, et al., Phosphoramidate Prodrugs of 2′-C-Methylcytidine for Therapy of Hepatitis C Virus Infection, Journal of Medicinal Chemistry, 2009, pp. 5394-5407, vol. 52, No. 17.
George et al., “Preparation of silyl-and germylmetallic compounds,” Journal of the American Chemical Society, Jan. 1960, 82(2):403-6.
Gleeson, et al., Prediction of the Potency of Inhibitors of Adenosine Deaminase by QM/MM Calculations, Chem. Commun., 2003, pp. 2180-2181.
Gordon, et al., Control of Hepatitis C: A Medicinal Chemistry Perspective, J. Med. Chem., 2005, pp. 1-20, vol. 48, No. 1.
Greene, et al., Protective Groups in Organic Synthesis, 1991, pp. 118-142, John Wiley & Sons.
Greene, T.W. and Wuts, P.G.M. (1991) Protective Groups in Organic Synthesis, published by John Wiley & Sons, v Inc., p. 1-4, 10-14, 47-53 and 100-103.
Grein et al., “Compassionate Use of Remdesivir for Patients with Severe Covid-19”, The New England Journal of Medicine, Apr. 2020, 382(24): 2327-2336.
Gudmundsson, et al., Synthesis of imidazo[1,2-a]pyridine C-Nucleosides with an Unexpected Site of Ribosylation, Journal of Organic Chemistry, 1997, pp. 3453-3459, vol. 62.
Gudmundsson, et al., The Condensation of 2,6-dichloroimidazo[1,2-a]pyridine C-nucleoside with an Unexpected Site of Ribosylation, Tetrahedron Letters, 1996, pp. 2365-2368, vol. 7, No. 14.
Gunic, et al., Cyclic monophosphate prodrugs of base-modified 2′-C-methyl ribonucleosides as potent inhibitors of hepatitis C virus RNA replication, Bioorganic & Medicinal Chemistry Letters, 2007, pp. 2452-2455, vol. 17.
Hamann, et al., Synthesis and antiviral evaluation of 7,9-dideaza-8-thiapurine C-nucleoside derivatives, Collection Symposium Series, 2008, pp. 347-349, vol. 10.
Hamann, et al., Synthesis and antiviral evaluation of thieno[3,4-d]pyrimidine C-nucleoside analogues of 2′,3′-dideoxy- and 2′,3′-dideoxy-2′,3′-didehydro-adenosine and -inosine, Bioorganic & Medicinal Chemistry, 2009, pp. 2321-2326, vol. 17.
Han, et al., Synthesis of 1-Chloroacetyl-1-dehydroxy-2,3,5-tri-O-benzoyl-β-D-ribofuranose. A Potentially Versatile Intermediate for the Synthesis of C-Nucleosides, Synthetic Communications, 1992, pp. 2815-2822, vol. 22, No. 19.
Haraguchi, K. et al., Stereoselective Synthesis of 1′-C-Branched Uracil Nucleosides From Uridine, Nucleosides & Nucleotides, 1995, pp. 417-420, vol. 14, No. 3-5.
Harcourt, et al., Molecular Characterization of the Polymerase Gene and Genomic Termini of Nipah Virus, Virology, 2001, pp. 192-201, vol. 287.
Harki, et al., Synthesis and Antiviral Activity of 5-Substituted Cytidine Analogues: Identification of Potent Inhibitor of Viral RNA-Dependent RNA Polymerases, Journal of Medicinal Chemistry, 2006, pp. 6166-6169, vol. 49, No. 21.
Hayashi, et al., C-Nucleosides, A Synthesis of 2-Substituted 7-(b-D-Ribofuranosyl)-Pyrrolo[2,1-f]-1,2,4-Triazines. A New Type of “Purine Like” C-Nucleoside, Heterocycles, 1992, pp. 569-574, vol. 34, No. 3.
Hecker, et al., Liver Targeted Prodrugs of 2′-C-Methyladenosine for Therapy of Hepatitis C Virus Infection, J. Med. Chem., 2007, pp. 3891-3896, vol. 50, No. 16.
Hoffmann, et al., When, in the context of drug design, can a fluorine atom successfully substitute a hydroxyl group?, International Journal of Quantum Chemistry, 2002, pp. 419-427, vol. 89.
Holshue et al., “First Case of 2019 Novel Coronavirus in the United States”, The New England Journal of Medicine, Jan. 2020, 9 pages.
Itoh, et al., Divergent and Stereocontrolled Approach to the Synthesis of Uracil Nucleosides Branched at the Anomeric Position, J. Org. Chem, 1995, pp. 656-662, vol. 60.
Jasko, et al., 5′-Phosphonates of Ribonucleosides and 2′-Deoxyribonucleosides: Synthesis and Antiviral Activity, Nucleosides & Nucleotides, 1993, pp. 879-893, vol. 12, No. 8.
Kabat et al., “Nucleosides, CXL VIII, Synthesis of 6-(β-D-Ribofuranosyl)picolinamide: A Novel C-Nucleoside from D-Ribonolactone”, Chemical & Pharmaceutical Bulletin, 1988, pp. 634-640, vol. 36, No. 2.
Kaewkhao et al., “High sensitivity methods to quantify chloroquine and its metabolite in human blood samples using LC-MS/MS”, Bioanalysis, Mar. 2019, 11(5):333-347.
Kalil et al., “Baricitinib plus Remdesivir for hospitalized adults with Covid-19,” New England Journal of Medicine, Dec. 11, 2020, 13 pages.
Khamnei, et al., Neighboring Group Catalysis in the Design of Nucleotide Prodrugs, J. Med. Chem., 1996, pp. 4109-4115, vol. 39, No. 20.
Kim, et al., Reversal of the Progression of Fatal Coronavirus Infection in Cats by a Broad-Spectrum Coronavirus Protease Inhibitor, PLOS Pathogens, Mar. 30, 2016, 18 pages.
Klumpp, et al., The Novel Nucleoside Analog R1479 (4′-Azidocytidine) is a Potent Inhibitor of NS5B-dependent RNA Synthesis and Hepatitis C virus Replication in Cell Culture, Journal of Biological Chemistry, 2006, pp. 3793-3799, vol. 281, No. 7.
Knaggs, et al., A QSAR Study Investigating the Effect of L-Alanine Ester Variation on the Anti-HIV Activity of Some Phosphoramidate Derivatives of d4T, 2000, pp. 2075-2078.
Knutsen, et al., Synthesis of Imidazo-fused Bridgehead-nitrogen C-Nucleosides : Coupling-Elimination Reactions of 2,5-Anhydro-3,4,6-tri-O-benzoyl-D-allonic Acid, J. Chem. Soc. Perkin Trans I, 1985, pp. 621-630.
Knutsen, et al., Synthesis of Imidazo-fused Bridgehead-nitrogen C-Nucleosides via Dehydrative Coupling Reactions of 2,5-Anhydro-3,4,6-tri-O-benzoyl-D-allonic Acid, J. Chem. Soc. Perkin Trans I, 1984, pp. 229-238.
Kobe, et al., Use of Distance Geometry Approach for the In Vitro Antiviral Activity Evaluation of N-bridgehead C-nucleosides, European J. Med. Chem., 1992, pp. 259-266, vol. 27, No. 3.
Kuzik et al., “Nebulized Hypertonic Saline in the Treatment of Viral Bronchiolitis in Infants”, The Journal of Pediatrics, Sep. 2007, 151(3):266-270.e1.
Lefebvre, et al., Mononucleoside Phosphotriester Derivatives with S-Acyl-2-thioethyl Bioreversible Phosphate-Protecting Groups: Intracellular Delivery of 3′-Azido-2′,3′-dideoxythymidine 5′-Monophosphate, Journal of Medicinal Chemistry, 1995, pp. 3941-3950, vol. 38, No. 20.
Lefebvre, et al., Synthesis, Decomposition Pathways and ‘In Vitro’ Evaluation of Bioreversible Phosphotriesters of Azt, Nucleosides, Nucleotides & Nucleic Acids, 1995, pp. 763-766, vol. 14, No. 3-5.
Lindell, et al., Synthesis and Biochemical Testing of 3-(Carboxyphenylethyl)imidazo[2,1- f][1,2,4]triazines as Inhibitors of AMP Deaminase, ACS Medicinal Chemistry Letters, 2010, pp. 286-289, vol. 1, No. 6.
Lo et al., “GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses,” Scientific Reports, 2017, 7(43395):1-7.
Lovelette, 1,2,4-Triazines. Synthesis of selected members of the s-triazolo[3,4-f][1,2,4]triazine and tetrazolo[1,5-f][1,2,4]triazine ring systems, Journal of Heterocyclic Chemistry, 1979, pp. 555-560, vol. 16.
Lu, Chengping, Veterinary Microbiology 5th edition, Jan. 31, 2013, p. 431, China Agriculure Press (No. English Translation available).
Martell, et al., Hepatitis C Virus (HCV) Circulates as a Population of Different but Closely Related Genomes: Quasispecies Nature of HCV Genome Distribution, Journal of Virology, 1992, pp. 3225-3229, vol. 6695.
Mason, et al., Polyadenylation-dependent screening assay for respiratory syncytial virus RNA transcriptase activity and identification of an inhibitor, Nucleic Acids Research, 2004, pp. 4758-4767, vol. 32, No. 16.
Matulic-Adamic, et al., Synthesis of 3-(β-D-Ribofuranosyl)-2-Fluoropyridine and 3-(β-D-Ribofuranosyl)-Pyridin-2-one, Tetrahedron Letters, 1997, pp. 203-206, vol. 38, No. 2.
Matulic-Adamic, et al., Synthesis of 5-β-D-Ribofuranosyl)-Pyridin-2-one: a ‘Deletion-Modified’ Analogue of Uridine, Tetrahedron Letters, 1997, pp. 1669-1672, vol. 38, No. 10.
McGuigan, et al. Application of Phosphoramidate ProTide Technology Significantly Improves Antiviral Potency of Carbocyclic Adenosine Derivatives, 2006, pp. 7215-7226.
McGuigan, et al., Aryl Phosphoramidate Derivatives of d4T Have Improved Anti-HIV Efficacy in Tissue Culture and May Act by the Generation of a Novel Intracellular Metabolite, J. Med. Chem., 1996, pp. 1748-1753, vol. 39.
McGuigan, et al., Intracellular Delivery of Bioactive AZT Nucleotides by Aryl Phosphate Derivatives of AZT, J. Med. Chem., 1993, pp. 1048-1052, vol. 36, No. 8.
Mehellou, et. al., Aryloxy Phosphoramidate Triesters: a Technology for Delivering Monophosphorylated Nucleosides and Sugarsinto Cells, ChemMedChem, 2009, pp. 1779-1791, vol. 4.
Meppen, et al., Cyclic phosphoramidates as prodrugs of 2′-C-methylcytidine, European Journal of Medicinal Chemistry, 2009, pp. 3765-3770, vol. 49, No. 9.
Meppen, et al., Medi-404—A Prodrug Approach for the Treatment of HCV Infection, Abstracts of papers, 236th ACS National Meeting, Philadelphia, PA, United States, Aug. 17-21, 2008.
Metobo, et al., Practical synthesis of 1 ′-substituted Tubercidin C-nucleoside analogs, Tetrahedron Letters, Feb. 2012, pp. 484-486, vol. 53, No. 5.
Migliaccio, et al., Characterization of Resistance to Non-obligate Chain-terminating Ribonucleoside Analogs That Inhibit Hepatitis C Virus Replication in vitro, The Journal of Biological Chemistry, 2003, pp. 49164-49170, vol. 278, No. 49.
Mitchell, et al., Bioreversible Protection for the Phospho Group: Bioactivation of the Di(4-acyloxybenzyl) and Mono(4-acyloxybenzyl) Phosphoesters of Methylphosphonate and Phosphonoacetate, J. Chem. Soc., Perkin Trans. 1, 1992, pp. 2345-2353.
Mitchell, et al., Synthesis of C-Nucleoside Isosteres of 9-(2-Hydroxyethoxymethyl)guanine (Acyclovir), J. Het. Chem., 1984, pp. 697-699, vol. 21, No. 3.
Moennig, et al., The Pestiviruses, Advances in Virus Research, 1992, pp. 53-98, vol. 41.
Moradpour, et al., Replication of hepatitis C virus, Nature Reviews Microbiology, 2007, pp. 453-463, vol. 5, No. 6.
Morris, “Mechanisms of action and therapeutic role of corticosteroids in asthma”, J. Allergy Clin. Immunol., Jan. 1985, 75(1 Pt):1-13.
Moscow, et al., Reduced Folate Carrier Gene (RFC1) Expression and Anti-Folate Resistance in Transfected and Non-Selected Cell Lines, International Journal of Cancer, 1997, pp. 184-190, vol. 72.
Mossel et al., “Exogenous ACE2 expression allows refractory cell lines to support severe acute respiratory syndrome coronavirus replication,” Journal of Virology, Mar. 15, 2005, 79(6):3846-50.
Munster et al., “Hydroxychloroquine concentration-response relationships in patients with rheumatoid arthritis”, Arthritis Rheumatology, Jun. 2002, 46(6):1460-1469.
Murakami et al., “Mechanism of Activation of Beta-D-2′-Fluoro-2′-C-Methylcytidine and Inhibition of Hepatitis C Virus NS5B RNA Polymerase”, Antimicrob Agents Chemother., Feb. 2007, 51(2):503-509.
Murakami et al., “Mechanism of Activation of PSI-7851 and Its Diastereoisomer PSI-7977”, The Journal of Biological Chemistry, 2010, 285(45):34337-34347.
Murphy et al., “The Nucleoside Analog GS-441524 Strongly Inhibits Feline Infectious Peritonisitis (FIP) Virus in Tissue Culture and Experimental Cat Infection Studies”, Veterinary Microbiology, 2018, 219:226-233.
Neumann et al., “Hepatitis C Viral Dynamics in Vivo and the Antiviral Efficacy of Interferon-α Therapy”, Science, 1998, 282:103-107.
Nishimura, et al., Synthesis of pyrrolo[2,1-f][1,2,4]triazine C-nucleosides. Isosteres of sangivamycin, tubercidin, and toyocamycin, Carbohydrate Research, 2001, pp. 77-82, vol. 331.
Ogura, et al., Reaction of Ethynyl Compounds with Lactones, Journal of Organic Chemistry, 1972, pp. 72-75, vol. 37, No. 1.
Olsen et al., “A 7-Deaza-Adenosine Analog Is a Potent and Selective Inhibitor of Hepatitis C Virus Replication with Excellent Pharmacokinetic Properties,” Antimicrobial agents and Chemotherapy, 2004, 3944-3953.
Otter, B. et al., Conformational Properties of Purine-Like C-Nucleosides, Nucleosides & Nucleotides, 1996, pp. 793-807, vol. 15, No. 1-3.
Pankiewicz, et al., C-Nucleoside Analogues of Nicotinamide Mononucleotide (NMN), Nucleosides and Nucleotides, 1988, pp. 589-593, vol. 7, No. 5&6.
Pankiewicz, et al., Efficient Synthesis of 5-(β-D-Ribofuranosyl)nicotinamide and its α-Isomer, Journal of Organic Chemistry, 1988, pp. 3473-3479, vol. 53.
Patani et al., “Bioisosterism: a rational approach in drug design,” Chem. Rev., 1996, 96:3147-3176.
Patil, et al., C-Glycosylation of Substituted Heterocycles under Friedel-Crafts Conditions (II): Robosylation of Multi-Functionalized Thiophenes and Furans for the Synthesis of Purine-Like C-Nucleosides, Nucleosides & Nucleotides, 1990, pp. 937-956, vol. 9, No. 7.
Patil, et al., Synthesis of Pyrrolo[2,1-f][1,2,4]triazine Congeners of Nucleic Acid Purines via the N-Amination of 2-Substituted Pyrroles, J. Het. Chem., 1994, pp. 781-786, vol. 31.
Patil, et al., Synthesis of some new thieno[3,4-d]pyrimidines and their C-nucleosides, Journal of Heterocyclic Chemistry, 1993, pp. 509-515, vol. 30, No. 2.
Patil, S. et al., 4-Aza-7,9-Dideazaadenosine, a New Cytotoxic Synthetic C-Nucleoside Analogue of Adenosine, Tetrahedron Letters, 1994, pp. 5339-5342, vol. 35, No. 30.
Perrone et al., “Application of the Phosphoramidate ProTide Approach to 4′-Azidouridine Confers Sub-micromolar Potency versus Hepatitis C Virus on an Inactive Nucleoside”, Journal of Medicinal Chemistry, 2007, 50(8):1840-1849.
Peterson, et al., Prodrug approaches to improving the oral absorption of antiviral nucleotide analogues, Expert Opinion, Drug Deliv., 2009, pp. 405-420, vol. 6, No. 4.
Piccirilli, et al., A Direct Route to 3-(D-Ribofuranosyl)pyridine Nucleosides, Helvetica Chimica Acta, 1991, pp. 397-406, vol. 74.
Pierra, et al., Synthesis and Pharmacokinetics of Valopicitabine (NM283), and Efficient Prodrug of the Potent Anti-HCV Agent 2′-C-Methylcytidine, Journal of Medicinal Chemistry, 2006, pp. 6614-6620, vol. 49, No. 22.
Poduch, et al., Design of Inhibitors of Orotidine Monophosphate Decarboxylase Using Bioisosteric Replacement and Determination of Inhibition Kinetics, Journal of Medicinal Chemistry, 2006, pp. 4937-4945, vol. 49, No. 16.
Porter, et al., Zika virus, drug discovery, and student projects, ScienceBlogs, Mar. 9, 2016, 7 pages.
Puech, et al., Intracellular Delivery of Nucleoside Monophosphates through a Reductase-mediated Activation Process, Antiviral Research, 1993, pp. 155-174, vol. 22, No. 4.
Ramasamy, et al., Synthesis and Antitumor Activity of Certain 3-B-D-Ribofuranosyl-1,2,4-triazolo[3,4-f]-1,2,4-triazines Related to Formycin Prepared via Ring Closure of a 1,2,4-Triazine Precursor, J. Med. Chem., 1986, pp. 2231-2235, vol. 29, No. 11.
Rao, et al., C-Glycosylation of Substituted Heterocycles under Friedel-Crafts Conditions (I): A Two-Step Synthesis of the Thieno[3,4-d]Pyrimidine C-Nucleoside Analog of Inosine, Tetrahedron Letters, 1988, pp. 3537-3540, vol. 29, No. 29.
Reddy, et al., Stereoselective Synthesis of Nucleoside Monophosphate HepDirectTM Prodrugs, Tet. Lett., 2005, pp. 4321-4324, vol. 46.
Ross, et al., Synthesis of Diastereomerically Pure Nucleotide andPhosphoramidates, J. Org. Chem., 2011, pp. 8311-8319, vol. 76.
Sacramento, et al., The clinically approved antiviral drug Sofosbuvir inhibits Zika Virus replication, Nature, Jan. 18, 2017.
Schul, et al., A Dengue Fever Viremia Model in Mice Shows Reduction in Viral Replication and Suppression of the Inflammatory Response after Treatment with Antiviral Drugs, Journal of Infectious Diseases, 2007, pp. 665-674, vol. 195.
Schultz, Prodrugs of Biologically Active Phosphate Esters, Bioorganic & Medicinal Chemistry, 2003, pp. 885-898, vol. 11.
Scott, et al., Interferon-a-2b Plus Ribavirin: A Review of its Use in the Management of Chronic Hepatitis C, Drugs, 2002, pp. 507-556, vol. 62, No. 3.
Sheahan et al., “Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses”, Science Translational Medicine, Jun. 2017, 9(396):eaal3653, 11 pages.
Sheahan, “Preparing for future pandemics, today with broad-spectrum antivirals”, Nature Portfolio Microbiology Community, Jan. 10, 2020, retrieved on May 13, 2021, retrieved from URL <“https://naturemicrobiologycommunity.nature.com/posts/58125-preparing-for-future-pandemics-today-with-broad-spectrum-antivirals”, 13 pages.
Shekunov, et al., Crystallization processes in pharmaceutical technology and drug delivery design, Journal of Crystal Growth, 2000, pp. 122-136, vol. 211.
Siegel et al., “Discovery and Synthesis of a Phosphoramidate Prodrug of a Pyrrolo[2,1-f][triazin-4-amino] Adenine C-Nucleoside(GS-5734) for the Treatment of Ebola and Emerging Viruses”, Journal of Medicinal Chemistry, 2017, 60(5): 1648-1661.
Siegel, et al., Discovery and Synthesis of a Phosphoramidate Prodrug of a Pyrrolo[2,1-f][triazin-4-amino] Adenine C-Nucleoside (GS-5734) for the Treatment of Ebola and Emerging Viruses, J. Med. Chem., 2017, 60, 5, 1648-1661 Supplementary Material.
Silverman et al., The Organic Chemistry of Drug Design and Drug Action, 1992, pp. 19-23.
Silverman, The Organic Chemistry of Drug Design and Drug Action, 2nd Ed., 2004, pp. 29-34.
Srivastav, et al., Antiviral Activity of Various 1-(2′-Deoxy-β-D-lyxofuranosyl), 1-(2′-Fluoro-β-D- ylofuranosyl), 1-(3′-Fluor-β-D-arabinofuranosyl), and 2′-Fluoro-2′,3′-didehydro-2′,3′-dideoxyribose Pyrimidine Nucleoside Analogues against Duck Hepatitis B Virus (DHBV) and Human Hepatitis B Virus (HBV) Replication, Journal of Medicinal Chemistry, 2010, pp. 7156-7166, vol. 53, No. 19.
Streetman, “Drug Interaction Concerns for COVID-19 Treatments”, Wolters Kluwer, Apr. 15, 2020, retrieved on Sep. 7, 2021, retrieved from URL <“https://www.wolterskluwer.com/en/expert-insights/drug-interaction-concerns-for-covid-19-treatments”>, 10 pages.
Sun, “Remdesivir for Treatment of COVID-19: Combination of Pulmonary and IV Administration May Offer Aditional Benefit”, The AAPS Journal, 2020, 22(77):1-6.
Tapia, et al., Combination of a Mutagenic Agent with a Reverse Transcriptase Inhibitor Results n Systematic Inhibition of HIV-1 Infection, Virology, 2005, pp. 1-8, vol. 338.
Totura et al., “Broad-spectrum coronavirus antiviral drug discovery”, Expert Opinion on Drug Discovery, Mar. 2019, 17 pages.
Towner, et Al., Newly Discovered Ebola Virus Associated with Hemorrhagic Fever Outbreak in Uganda, PLoS Pathogens, 2008, 6 pages, vol. 4, Issue 11.
Uchiyama, et al., O-selective Phosphorylation of Nucleosides without N-protection, J. Org. Chem., Jan. 1, 1993, vol. 58, No. 2.
Vaghefi, et al., Synthesis and Antiviral Activity of Certain Nucleoside 5′-Phosphonoformate Derivatives, Journal of Medicinal Chemistry, 1986, pp. 1389-1393, vol. 29, No. 8.
Venkatachalam, et al. Effect of change in nucleoside structure on the activation and antiviral activity of phosphoramidate derivatives, 2005, pp. 5408-5423.
Walker et al., “Plasma chloroquine and desethylchloroquine concentrations in children during and after chloroquine treatment for malaria.”, British Journal Clinical Pharmacology, Dec. 1983, 16(6):701-705.
Wang et al., “Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro”, Cell Research, 2020, 30:269-271.
Warren et al., “Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430”, Nature, Apr. 2014, 508(7496):402-405.
Warren et al., “Therapeutic efficacy of the small molecules GS-5734 against Ebola virus in rhesus monkeys”, Nature, Mar. 17, 2016, 19 pages.
Wu, et al., Synthetic Methodologies for C-Nucleosides, Synthesis, 2004, pp. 1533-1553, vol. 10.
Xie et al., “A nanoluciferase SARS-CoV-2 for rapid neutralization testing and screening of anti-infective drugs for COVID-19,” Nature Communications, Oct. 15, 2020, 11(1):1-1.
Yamanaka, et al., Metabolic Studies on BMS-200475, a New Antiviral Compound Active against Hepatitis B Virus, Antimicrobial Agents and Chemotherapy, 1999, p. 190, vol. 43, No. 1.
Yates et al., “The evolution of antiviral nucleoside analogues: A review for chemists and non-chemists. Part II: Complex modifications to the nucleoside scaffold”, Antiviral Research, Dec. 8, 2018, 162:5-21.
Yoshimura, et al., Synthesis and Biological Evaluation of 1′-C-Cyano-Pyrimidine Nucleosides, Nucleosides & Nucleotides, 1996, pp. 305-324, vol. 15, No. 1-3.
Zhang et al., “A Practical Synthesis of (2R)-3,5-di-O-benzoyl-2-fluoro-2-C-methyl-D-ribono-y-lactone”, Tetrahedron: Asymmetry, 2009, 20:305-312.
Zhu et al., “A novel coronavirus from patients with pneumonia in China, 2019,” New England Journal of Medicine, Jan. 24, 2020, 14 pages.
Aripo Patent Office, Form 21 and Substantive Examination Report (in English) for AP Application No. AP/P/2010/005439, dated Mar. 18, 2014.
Aripo Patent Office, Form 21 for AP Patent Application No. AP/P/2011/005818, Sep. 19, 2013.
Aripo Patent Office, Search and Exam Report for AP Application No. AP/P/2012/006189, dated Jun. 26, 2014.
Aripo Patent Office, Search Report for AP Patent Application No. AP/P/2011/005818, dated Sep. 19, 2013.
Australia Patent Office, First Examination Report for AU Patent Application No. 2009240630, dated Jun. 14, 2012.
Australia Patent Office, Patent Examination Report No. 1 for AU Application No. 2011306066, dated Nov. 21, 2013.
Australia Patent Office, Patent Examination Report No. 1 for AU Patent Application No. 2010213873, dated Jun. 4, 2014.
Chile Patent Office, First Office Action for CL Patent Application No. 1906-2011, dated May 7, 2013.
Chile Patent Office, Opposition for CL Patent Application No. 727-2013, Oct. 15, 2013.
Chile Patent Office, Second Office Action for CL Patent Application No. 1906-2011, dated Oct. 16, 2013.
Chinese Patent Office, First Examination Report for CN Patent Application No. 200980120218.8, dated Nov. 13, 2012.
Chinese Patent Office, First Office Action for CN Patent Application No. 201080011690.0, dated Jun. 8, 2013.
Chinese Patent Office, Notification of Reexamination for CN Patent Application No. 200980120218.8, dated Sep. 1, 2014.
Chinese Patent Office, Notification of the Second Office Action & Search Report for CN Patent Application No. 201080011690.0, dated Jan. 8, 2014.
Chinese Patent Office, Notification of the Third Office Action for CN Patent Application No. 201080011690.0, dated Jul. 29, 2014.
Chinese Patent Office, Rejection Decision for CN Patent Application No. 200980120218.8, dated Feb. 7, 2014.
Chinese Patent Office, Second Examination Report for CN Patent Application No. 200980120218.8, dated Jun. 21, 2013.
Columbia Patent Office, First Examination Report (in English) for CO Patent Application No. 10-131479, dated Oct. 23, 2012.
Columbia Patent Office, Office Action for CO Patent Application No. 11-109.501, dated Nov. 27, 2012.
Columbia Patent Office, Office Action for CO Patent Application No. 13-235103-1, dated Aug. 27, 2014.
Columbia Patent Office, Resolution No. 56673 for CO Patent Application No. 10-131479, Sep. 27, 2013.
Columbia Patent Office, Second Examination Report (in English) for CO Patent Application No. 10-131479, dated Jun. 20, 2013.
Columbian Patent Office, Office Action No. 425 for CO Patent Application No. 12 050 579, dated Jan. 21, 2014.
Ecuador Patent Office, Opposition for EC Patent Application No. SP-2012-11817, May 27, 2013.
El Salvador Patent Office, Official Action for SV National Phase Entry of International Application No. PCT/US2010/049471, dated Nov. 6, 2013.
Eurasian Patent Office, First Examination Report for EA Patent Application No. 201071128, dated Apr. 25, 2012.
Eurasian Patent Office, First Office Action for EA Patent Application No. 201190110/28, dated Apr. 26, 2012.
Eurasian Patent Office, First Office Action for EA Patent Application No. 201390141/28, with English translation, dated Aug. 14, 2014.
Eurasian Patent Office, Office Action for EA Patent Application No. 201390152, dated Apr. 14, 2014.
Eurasian Patent Office, Official Action for EA Patent Application No. 201390133, dated Mar. 27, 2014.
Eurasian Patent Office, Second Examination Report for EA Patent Application No. 201071128, dated Oct. 24, 2012.
Eurasian Patent Office, Second Office Action for EA Patent Application No. 201190110/28, dated Jan. 28, 2013.
Eurasian Patent Office, Third Examination Report for EA Patent Application No. 201071128, dated Apr. 29, 2013.
Eurasian Patent Office, Third Office Action for EA Application No. 201190110/28, dated Oct. 18, 2013.
European Patent Office, Communication pursuant to Article 94(3) EPC for EP Patent Application No. 10763083.2, dated May 2, 2014.
European Patent Office, Communication pursuant to Article 94(3) EPC for EP Patent Application No. 11715792.5, dated Feb. 14, 2014.
European Patent Office, Communication under 161/162 for EP Patent Application No. 10704068.5, dated Sep. 6, 2011.
European Patent Office, Communication under 161/162 for EP Patent Application No. 10763083.2, dated May 11, 2012.
European Patent Office, Communication under 161/162 for EP Patent Application No. 11715792.5, dated Apr. 26, 2013.
European Patent Office, First Office Action for EP Patent Application No. 10704068.5, dated Jun. 18, 2012.
Indonesia Patent Office, First Examination Report for ID Patent Application No. W00 2010 03957, dated Apr. 25, 2013.
Indonesia Patent Office, Substantive Examination Report Stage 1 for ID Application No. W-00201103126, dated Jun. 10, 2014.
Israel Patent Office, First Examination Report for IL Patent Application No. 208701, dated Jan. 13, 2013.
Israel Patent Office, First Office Action for IL Patent Application No. 214396, dated Jul. 8, 2013.
Israel Patent Office, Notification of Defects for IL Patent Application No. 214396, dated Nov. 11, 2013.
Israel Patent Office, Notification of Defects for IL Patent Application No. 218599, dated Aug. 25, 2014.
Israel Patent Office, Notification of Defects for IL Patent Applicaton No. 208701, dated Aug. 25, 2014.
Israel Patent Office, Notification Prior to Examination for IL Patent Application No. 218599, dated Nov. 13, 2012.
Japanese Patent Office, First Examination Report for JP Patent Application No. 2011-506429, dated Aug. 22, 2013.
Japanese Patent Office, Notice of Reasons for Rejection for Japanese Patent Appln. No. JP 2017-520934, dated Mar. 30, 2018.
Japanese Patent Office, Notification of Reasons for Rejection for JP Application No. 2011-549324, dated Jul. 28, 2014.
Japanese Patent Office, Notification of Reasons for Rejection for JP Application No. 2011-549324, dated Mar. 26, 2014.
Japanese Patent Office, Notification of Reasons for Rejection for JP Patent Application No. 2012-529958, dated Aug. 5, 2014.
Mexico Patent Office, English translation of Office Action for MX Application No. MX/a/2013/003179, dated Feb. 25, 2014.
Mexico Patent Office, First Examination Report (in English) for MX Patent Application No. MX/a/2010/011661, dated Oct. 26, 2011.
Mexico Patent Office, Office Action for MX Application No. MX/a/2011/008409, dated Mar. 25, 2014.
New Zealand Patent Office, First Examination Report for NZ Patent Application No. 588670, dated Apr. 8, 2011.
New Zealand Patent Office, First Examination Report for NZ Patent Application No. 608070, dated Nov. 7, 2013.
New Zealand Patent Office, Further Examination Report for NZ Application No. 594370, dated Oct. 8, 2013.
PCT International Preliminary Report on Patentability and Written Opinion for PCT International Application No. PCT/US2009/041432, dated Oct. 26, 2010, 7 pages.
PCT International Preliminary Report on Patentability and Written Opinion for PCT International Application No. PCT/US2010/049471, dated Mar. 27, 2012, 7 pages.
PCT International Preliminary Report on Patentability and Written Opinion for PCT International Application No. PCT/US2011/038253, dated Dec. 4, 2012, 6 pages.
PCT International Preliminary Report on Patentability for PCT International Application No. PCT/US2010/023586, dated Aug. 16, 2011, 6 pages.
PCT International Preliminary Report on Patentability for PCT International Application No. PCT/US2011/028897, dated Mar. 26, 2013, 7 pages.
PCT International Preliminary Report on Patentability for PCT International Application No. PCT/US2011/029441, dated Mar. 26, 2013, 7 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2009/041432, dated Aug. 11, 2009, 11 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2011/028897, dated Aug. 1, 2011, 6 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2011/029441, dated Aug. 1, 2011, 5 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2011/038253, dated Jul. 29, 2011, 4 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2017/028251, dated Oct. 16, 2017, 22 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2017/028251, dated Sep. 13, 2017, 22 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2010/023586, dated Aug. 4, 2010, 4 pages.
PCT International Search Report and Written Opinion for PCT International Application No. PCT/US2010/049471, dated Nov. 18, 2010, 11 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2021/034764, dated Aug. 23, 2021, 16 pages.
Peru Patent Office, Office Action in PE Application No. 1464, dated Sep. 12, 2013.
Resolution No. 48031 for CO Patent Application No. 10-121.513, rec'd Oct. 7, 2014 (8 pages) (English translation).
Ukraine Patent Office, First Examination Report for UA Patent Application No. 2010 13030, dated Mar. 2, 2013.
Ukraine Patent Office, First Office Action for UA Application No. a 2011 10568, dated Apr. 7, 2014.
Ukraine Patent Office, Second Office Action for UA Patent Application No. 2011 10568, dated Aug. 11, 2014.
Vietnam Patent Office, First Examination Report for VN Patent Application No. 1-2010-02939, dated Apr. 19, 2012.
Vietnam Patent Office, Second Examination Report for VN Patent Application No. 1-2010-02939, dated Jul. 26, 2012.
Adlington et al., “Synthesis of novel C-nucleosides with potential applications in combination and parallel synthesis,” Tetrahedron Letters, 2000, 41:575-578.
Behzadi et al., “Overview of Current Therapeutics and Novel Candidates Against Influenza Respiratory Syncytial Virus, and Middle East Respiratory Syndrome Coronavirus Infections,” Frontiers in Microbiology, Jun. 2019, 10:1327, pp. 1-16.
Huang et al., “Recent development of therapeutics for chronic HCV infection,” Antiviral Research, Sep. 2006, 71(2-3): 351-362.
Kushner et al., “Pharmacological uses and perspectives of heavy water and deuterated compounds,” Canadian Journal of Physiology and Pharmacology, Feb. 1999, 77(2):79-88.
Martin et al., “Hint2, A Mitochondrial Apoptotic Sensitizer Down-Regulated in Hepatocellular Carcinoma,” Gastroenterology, Jun. 2006, 130(7):2179-2188.
Sahakijpijarn et al., “Development of Remdesivir as a Dry Powder for Inhalation by Thin Film Freezing,” Pharmaceutics, Oct. 2020, 12(11):1002, 27 pages.
Schnirring, “China releases genetic data on new coronavirus, now deadly,” CIDRAP News, Jan. 2020, retrieved on Mar. 15, 2022, retrieved from URL <https://www.cidrap.umn.edu/news-perspective/2020/01/china-releases-genetic-data-new-coronavirus-now-deadly>, 3 pages.
Shi et al., “Synthesis and anti-viral activity of a series of d- and 1-2′-deoxy-2′-fluororibonucleosides in the subgenomic HCV replicon system,” Bioorganic & Medicinal Chemistry, Mar. 2005, 13(5):1641- 1652.
Sofia et al., “Discovery of a β-d-2′-Deoxy-2′-a-fluoro-2′-β-C-methyluridine Nucleotide Prodrug (PSI-7977) for the Treatment of Hepatitis C Virus,” Journal of Medicinal Chemistry, Sep. 2010, 53(19):7202-7218.
Stein et al., “Phosphorylation of Nucleoside Analog Antiretrovirals: A Review for Clinicians,” Pharmacotherapy, Jan. 2001, 21(1):11-34.
Stella et al., “Cyclodextrins,” Toxicologic Pathology, 2008, 36(1):30-42.
Szente et al., “Sulfobutylether-beta-cyclodextrin-enabled antiviral remdesivir: Characterization of electrospun- and lyophilized formulations,” Carbohydrate Polymers, 2021, 264:118011, 8 pages.
Xie et al., “Weinreb Amide Approach to the Practical Synthesis of a Key Remdesivir Intermediate,” The Journal of Organic Chemistry, 2021, 86:5065-5072.
Bowie et al., “RIG-I: tri-ing to discriminate between self and non-self RNA,” Trends in Immunology, Apr. 2007, 28(4): 147-150.
Cox et al., “Therapeutically administered ribonucleoside analogue MK-4482/EIDD-2801 blocks SARS-CoV-2 transmission in ferrets,” Nature Microbiology, 2020, 6(1): 11-18.
Dinnon et al., “A mouse-adapted model of SARS-C.V-2 to test COVID-19 countermeasures,” Nature, Aug. 2020, 586: 560-566.
Flint et al., “Functional analysis of cell surface-expressed hepatitis C virus E2 glycoprotein,” J. Virol., Aug. 1999, 73(8): 6782-6790.
Freeman et al., “3 Prodrug Design for Phosphates and Phosphonates,” Progress in Medicinal Chemistry, 1997, 34: 111-147.
Pelet et al., “High throughput screening assay for negative single stranded RNA virus polymerase inhibitors,” J. Virol. Methods, Sep. 2005, 128(1-2): 29-36.
Perrone et al., “First Example of Phosphoramidate Approach Applied to a 4′-Substituted Purine Nucleoside (4′-Azidoadenosine): Conversion of an Inactive Nucleoside to a Submicromolar Compound versus Hepatitis C Virus,” Journal of Medicinal Chemistry, Oct. 2007, 50(22): 5463-5470.
Wang et al., “ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data,” Nucleic Acids Research, 2010, 38(16): e164, 7 pages.
Wolfel et al., “Virological assessment of hospitalized patients with COVID-2019,” Nature, Apr. 2020, 581: 465-470.
Yoon et al., “High-throughput screening-based identification of paramyxovirus inhibitors,” J. Biomol. Screen., Aug. 2008, 13(7): 591-608.
“Molecular Nuclear Medicine,” First Edition, Wang (ed.), May 31, 2001, pp. 388-391, 11 pages (with English translation).
“Veterinary Microbiology,” 4th Edition, Lu (ed.), 2007, p. 304: paragraph 2, p. 408: paragraph 1, p. 419: paragraphs 1-2, 7 pages (with English translation).
Aleissa et al., “New Perspectives on Antimicrobial Agents: Remdesivir Treatment for COVID-19,” Antimicrobial Agents and Chemotherapy, Dec. 2020, 65(1): 18 pages.
Anderson et al., “The use of convalescent plasma therapy and remdesivir in the successful management of a critically ill obstetric patient with novel coronavirus 2019 infection: A case report,” Case Reports in Women's Health, May 2020, 27: 3 pages.
Baker et al., “Prodrugs of 9-Beta-D-Arabinofuranosyladenine. 1. Synthesis and Evaluation of some 5′- (O-Acyl) Derivatives,” Journal of Medicinal Chemistry, Dec. 1978, 21(12): 1218-1221.
Beigel et al., “Remdesivir for the Treatment of Covid-19—Final Report,” New England Journal of Medicine, Nov. 5, 2020, 383(19): 1813-1826.
Chinen et al., “Critical respiratory failure in pregnancy complicated with COVID-19: A case report.” Case Reports in Women's Health, Apr. 2021, 30: 4 pages.
Dande et al., “Remdesivir in a pregnant patient with COVID-19 pneumonia,” Journal of Community Hospital Internal Medicine Perspectives, Jan. 2021, 11(1): 103-6.
De Wit et al., “Prophylactic and Therapeutic Remdesivir (GS-5734) Treatment in the Rhesus Macaque Model of MERS-CoV Infection,” Proceedings of the National Academy of Sciences of the United States of America, Mar. 2020, 117(12): 6771-6776.
Douafer et al., “Scope and limitations on aerosol drug delivery for the treatment of infectious respiratory diseases,” Journal of Controlled Release, Sep. 2020, 325: 276-292.
Easterlin et al., “Extremely Preterm Infant Born to a Mother With Severe COVID-19 Pneumonia,” Journal of Investigative Medicine High Impact Case Reports, Jul. 2020, 8: 1-5.
Gil et al., “COVID-19: Drug Targets and Potential Treatments,” Journal of Medicinal Chemistry, Jun. 2020. 63(21): 12359-12386.
He et al., Species Differences in Size Discrimination in the Paracellular Pathway Reflected by Oral Bioavailability of Poly(ethylene glycol) and D-peptides, Journal of Pharmaceutical Sciences, May 1998, 87(5): 626-633.
Humeniuk et al., “Pharmacokinetic, Pharmacodynamic, and Drug-Interaction Profile of Remdesivir, a SARS-CoV-2 Replication Inhibitor,” Clinical pharmacokinetics, May 2021, 60(2021): 569-583.
Igbinosa et al., “Use of remdesivir for pregnant patients with severe novel coronavirus disease 2019,” American Journal of Obstetrics & Gynecology, Aug. 2020, 223(5): 768-770.
Jacobson et al., “Use of dexamethasone, remdesivir, convalescent plasma and prone positioning in the treatment of severe COVID-19 infection in pregnancy: A case report,” Case Reports in Women's Health, Jan. 2021, 29: 3 pages.
Lat et al., “Therapeutic options in the treatment of severe acute respiratory syndrome coronavirus 2 in pregnant patient,” American Journal of Obstetrics & Gynecology MFM, Nov. 2020, 2(4): 100224.
Mackman et al., “Prodrugs of a 1′-CN-4-Aza-7,9-dideazaadenosine C-Nucleoside Leading to the Discovery of Remdesivir (GS-5734) as a Potent Inhibitor of Respiratory Syncytial Virus with Efficacy in the African Green Monkey Model of RSV,” Journal of Medicinal Chemistry, Apr. 2021, 64(8): 5001-5017.
Maldarelli et al., “Remdesivir Treatment for Severe COVID-19 in Third-Trimester Pregnancy: Case Report and Management Discussion,” Open Forum Infectious Diseases, Sep. 2020, 7(9): 4 pages.
Martin et al., “Genetic Conservation of SARS-CoV-2 RNA Replication Complex in Globally Circulating Isolates and Recently Emerged Variants from Humans and Minks Suggests Minimal Pre-Existing Resistance to Remdesivir,” Antiviral Research, Apr. 2021, 188: 7 pages.
McCoy et al., “Compassionate use of remdesivir for treatment of severe coronavirus disease 2019 in pregnant women at a United States academic center,” American Journal of Obstetrics & Gynecology MFM, Aug. 2020, 2(Suppl 3): 4 pages.
Mulangu et al., “A Randomized, Controlled Trial of Ebola Virus Disease Therapeutics,” New England Journal of Medicine, Dec. 2019; 381(24): 2293-303.
Naqvi et al., “Tocilizumab and Remdesivir in a Pregnant Patient With Coronavirus Disease 2019 (COVID-19),” Obstetrics & Gynecology, Nov. 2020, 136(5): 1025-9.
Nasrallah et al., “Pharmacological treatment in pregnant women with moderate symptoms of coronavirus disease 2019 (COVID-19) pneumonia,” The Journal of Maternal-Fetal & Neonatal Medicine Mar. 2021, 35(25): 5970-5977.
Pierce-Williams et al., “Clinical course of severe and critical coronavirus disease 2019 in hospitalized pregnancies: a United States cohort study,” American Journal of Obstetrics & Gynecology MFM, Aug. 2020, 2(3): 12 pages.
Pilcer et al., “Formulation strategy and use of excipients in pulmonary drug delivery,” International Journal of Pharmaceutics, Jun. 2010, 392(1-2): 1-19.
Saroyo et al., “Remdesivir Treatment for COVID 19 in Pregnant Patients with Moderate to Severe Symptoms: Serial Case Report,” Infectious Disease Reports, May 2021, 13(2): 437-443.
Schnettler et al., “Severe acute respiratory distress syndrome in coronavirus disease 2019-infected pregnancy: obstetric and intensive care considerations,” American Journal of Obstetrics & Gynecology MFM, Aug. 2020, 2(Suppl 3): 10 pages.
Singh et al., “Treatment With Remdesivir in Two Pregnant Patients With COVID-19 Pneumonia,” Cureus, May 2021, 13(5): 6 pages.
Spinner et al., “Effect of Remdesivir vs Standard Care on Clinical Status at 11 Days in Patients With Moderate COVID-19: A Randomized Clinical Trial.” Jama, Sep. 2020, 324(11): 1048-1057.
Taylor et al., “Neutralizing Monoclonal Antibodies for Treatment of COVID-19,” Nature Reviews Immunology, Apr. 2021, 21(6): 382-393.
Taylor, “Aulton's Pharmaceutics: The Design and Manufacture of Medicines; Chapter 37: Pulmonary Drug Delivery,” 5th ed., Aulton et al (ed), 2018: 653-670.
V'kovski et al., “Coronavirus Biology and Replication: Implications for SARS-CoV-2,” Nature Reviews Microbiology, Oct. 2021, 19(3): 155-170.
Xie et al., “Engineering SARS-CoV-2 using a reverse genetic system,” Nature protocols, Jan. 29, 2021, 16(3): 1761-1784.
Taiwanese Office Action in TW Appln. No. 110119391, dated Mar. 25, 2023, 12 pages (with English translation).
Office Action in Chinese Appln. No. 202180037821.0, mailed on Sep. 22, 2023, 12 pages (with English translation).
Office Action in European Appln. No. 21733348.3 mailed on Oct. 24, 2023, 6 pages.
“Medical Microbology,” Fourt Edition, Baron (ed.), University of Texas Medical Branch at Galveston, 1996, Chapter 59 and 72, 38 pages.
Beaucourt et al., “Ribaviring: a drug active against many viruses with multiple effects on virus replication and progragation. Molecular basis of ribavirin resistance,” Current Opinions in Virology, May 2014, 8:10-15.
Choe et al., “Exploration for the effect of renal function and renal replacement therapy on pharmacokinetics of remdesivir and GS—441524 in patients with COVID-19: A limited case series,” Clinical and Translational Science, Nov. 20, 2021, 15(3):732-740.
Japanese Office Action in Appln. No. 2022-573334, mailed Dec. 6, 2023, 8 pages (with English translation).
McGuigan et al., “Application of Phosporamidate Pronucleotide Technolgy to Abacavir Leads to a Significant Enhancement of Antiviral Potency”, J. Med. Chem. 2005, 48(10):3504-3515.
Office Action in Australian Appln. No. 2021281351, dated Oct. 26, 2023, 4 pages.
Ssentongo et al., BMC Infectious Diseases, “SARS-CoV-2 vaccine effectiveness against infection, symptomatic and severe COVID-19: a systematic review and meat-analysis”, vol. 22, Article 439, 2022. (Year: 2022).
U.S. Appl. No. 13/189,373, filed Jul. 22, 2011, Richard L. Mackman.
U.S. Appl. No. 14/613,719, filed Feb. 4, 2015, Richard L. Mackman.
U.S. Appl. No. 14/579,348, filed Dec. 22, 2014, Richard L. Mackman.
U.S. Appl. No. 16/042,085, filed Jul. 23, 2018, Richard L. Mackman.
U.S. Appl. No. 16/879,491, filed May 20, 2020, Richard L. Mackman.
U.S. Appl. No. 17/854,818, filed Jun. 30, 2022, Richard L. Mackman.
U.S. Appl. No. 17/333,389, filed May 28, 2021, Tomas Cihlar.
U.S. Appl. No. 17/676,920, filed Feb. 22, 2022, Tomas Cihlar.
U.S. Appl. No. 18/128,850, filed Mar. 30, 2023, Tomas Cihlar.
U.S. Appl. No. 18/540,002, filed Dec. 14, 2023, Tomas Cihlar.
U.S. Appl. No. 17/222,125, filed Apr. 5, 2021, Scott Ellis.
U.S. Appl. No. 18/202,751, filed Jun. 26, 2023, Scott Ellis.
U.S. Appl. No. 17/158,391, filed Jan. 26, 2021, Tomas Cihlar.
U.S. Appl. No. 18/131,106, filed Apr. 5, 2023, Tomas Cihlar.
U.S. Appl. No. 17/198,829, filed Mar. 11, 2021, Pavel R. Badalov.
U.S. Appl. No. 18/108,480, filed Feb. 10, 2023, Pavel R. Badalov.
U.S. Appl. No. 16/031,620, filed Jul. 10, 2018, Nate Larson.
U.S. Appl. No. 16/865,209, filed May 1, 2020, Nate Larson.
U.S. Appl. No. 17/585,651, filed Jan. 27, 2022, Nate Larson.
U.S. Appl. No. 18/241,303, filed Sep. 1, 2023, Nate Larson.
U.S. Appl. No. 15/919,750, filed Mar. 13, 2018, Michel Joseph Perron.
U.S. Appl. No. 16/852,102, filed Apr. 17, 2020, Michel Joseph Perron.
U.S. Appl. No. 17/578,682, filed Jan. 19, 2022, Michel Joseph Perron.
U.S. Appl. No. 17/895,123, filed Aug. 25, 2022, Michel Joseph Perron.
U.S. Appl. No. 18/133,612, filed Apr. 12, 2023, Michel Joseph Perron.
U.S. Appl. No. 18/519,194, filed Nov. 27, 2023, Michel Joseph Perron.
U.S. Appl. No. 15/964,597, filed Apr. 27, 2018, Katrien Brak.
U.S. Appl. No. 17/069,248, filed Oct. 13, 2020, Katrien Brak.
U.S. Appl. No. 18/099,477, filed Jan. 20, 2023, Katrien Brak.
U.S. Appl. No. 15/267,433, filed Sep. 16, 2016, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 16/265,016, filed Feb. 1, 2019, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 16/863,566, filed Apr. 30, 2020, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 17/222,066, filed Apr. 5, 2021, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 17/748,400, filed May 19, 2022, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 18/402,949, filed Apr. 3, 2024, Michael O'Neil Hanrahan Clarke.
U.S. Appl. No. 14/926,063, filed Oct. 29, 2015, Steven Donald Axt.
U.S. Appl. No. 16/692,966, filed Nov. 22, 2019, Steven Axt.
U.S. Appl. No. 17/665,724, Feb. 7, 2022, Steven Donald Axt.
U.S. Appl. No. 14/926,062, filed Oct. 29, 2015, Byoung Chun.
U.S. Appl. No. 15/246,240, filed Aug. 24, 2016, Byoung Chun.
U.S. Appl. No. 15/902,690, filed Feb. 22, 2018, Byoung Chun.
U.S. Appl. No. 16/274,049, filed Feb. 12, 2019, Byoung Chun.
U.S. Appl. No. 16/881,419, filed May 22, 2020, Byoung-Kwon Chun.
U.S. Appl. No. 17/579,650, filed Jan. 20, 2022, Byoung Kwon Chun.
U.S. Appl. No. 17/897,380, filed Aug. 29, 2022, Byoung Kwon Chun.
U.S. Appl. No. 18/134,792, filed Apr. 14, 2023, Byoung Kwon Chun.
U.S. Appl. No. 18/523,984, filed Nov. 30, 2023, Byoung Kwon Chun.
U.S. Appl. No. 13/813,886, filed Jun. 25, 2013, Aesop Cho.
U.S. Appl. No. 14/746,430, Jun. 22, 2015, Aesop Cho.
U.S. Appl. No. 12/886,248, filed Sep. 20, 2010, Thomas Butler.
U.S. Appl. No. 16/011,055, filed Jun. 18, 2018, Thomas Butler.
U.S. Appl. No. 16/988, 250, filed Aug. 7, 2020, Thomas Butler.
U.S. Appl. No. 17/209,639, filed Mar. 23, 2021, Thomas Butler.
U.S. Appl. no. 12/428,176, filed Apr. 22, 2009, Thomas Butler.
U.S. Appl. No 13/196,117, filed Aug. 2, 2011, Thomas Butler.
U.S. Appl. No. 13/649,511, filed Oct. 11, 2012, Thomas Butler.
U.S. Appl. No. 18/286,971, filed Oct. 13, 2023, Stacey Bremner.
U.S. Appl. No. 17/458,023, filed Aug. 26, 2021, Elaine Bunyan.
U.S. Appl. No. 18/098,950, filed Jan. 19, 2023, Elaine Bunyan.
U.S. Appl. No. 18/410,236, filed Jan. 11, 2023, Elaine Bunyan.
U.S. Appl. No. 18/115,895, filed Mar. 1, 2023, Rao V. Kalla.
U.S. Appl. No. 18/115,955, filed Mar. 1, 2023, Mark J. Bartlett.
U.S. Appl. No. 18/117,858, filed Mar. 6, 2023, Mark. J. Bartlett.
U.S. Appl. No. 18/117,878, filed Mar. 6, 2023, Mark J. Bartlett.
U.S. Appl. No. 18/117,913, filed Mar. 6, 2023, Mark J. Bartlett.
U.S. Appl. No. 18/237,152, filed Aug. 25, 2023, Mark J. Bartlett.
U.S. Appl. No. 17/355,813, filed Jun. 23, 2021, Daniel H. Byun.
U.S. Appl. No. 18/544,561, filed Dec. 19, 2023, Daniel H. Byun.
U.S. Appl. No. 18/205,745, filed Jun. 5, 2023, Roy Maxim Bannister.
U.S. Appl. No. 18/243,812, filed Sep. 8, 2023, Casey B. Davis.
U.S. Appl. No. 18/394,488, filed Dec. 22, 2023, Casey B. Davis.
U.S. Appl. No. 18/512,088, filed Nov. 17, 2023, John Philip Bilello.
U.S. Appl. No. 18/215,881, filed Jun. 6, 2023, filed Kassibla E. Dempah.
U.S. Appl. No. 18/384,060, filed Oct. 26, 2023, Kimberly T. Barrett.
U.S. Appl. No. 18/215,217, filed Jun. 28, 2023, Kassibla E. Dempah.
Related Publications (1)
Number Date Country
20210393653 A1 Dec 2021 US
Provisional Applications (1)
Number Date Country
63032321 May 2020 US