Treating sickle cell disease with a pyruvate kinase R activating compound

Information

  • Patent Grant
  • 11980611
  • Patent Number
    11,980,611
  • Date Filed
    Thursday, December 22, 2022
    a year ago
  • Date Issued
    Tuesday, May 14, 2024
    18 days ago
Abstract
Compounds that activate pyruvate kinase R can be used for the treatment of sickle cell disease (SCD). Methods and compositions for the treatment of SCD are provided herein, including a therapeutic compound designated as Compound 1.
Description
TECHNICAL FIELD

This disclosure relates to the treatment of sickle cell disease (SCD), including the treatment of patients diagnosed with SCD by the administration of a compound that activates pyruvate kinase R (PKR).


BACKGROUND

Sickle cell disease (SCD) is a chronic hemolytic anemia caused by inheritance of a mutated form of hemoglobin (Hgb), sickle Hgb (HgbS). It is the most common inherited hemolytic anemia, affecting 70,000 to 80,000 patients in the United States (US). SCD is characterized by polymerization of HgbS in red blood cells (RBCs) when HgbS is in the deoxygenated state (deoxy-HgbS), resulting in a sickle-shaped deformation. Sickled cells aggregate in capillaries precipitating vaso-occlusive events that generally present as acute and painful crises resulting in tissue ischemia, infarction, and long-term tissue damage. RBCs in patients with SCD tend to be fragile due to sickling and other factors, and the mechanical trauma of circulation causes hemolysis and chronic anemia. Finally, damaged RBCs have abnormal surfaces that adhere to and damage vascular endothelium, provoking a proliferative/inflammatory response that underlies large-vessel stroke and potentially pulmonary-artery hypertension. Collectively, these contribute to the significant morbidity and increased mortality associated with this disease.


Currently, therapeutic treatment of SCD is inadequate. The only known cure for SCD is hematopoietic stem cell transplantation which has serious risks, is typically recommended for only the most serious cases, and is largely offered only to children with sibling-matched donors. Gene therapy is also under investigation with promising preliminary results; however, there are market access hurdles, mainly high cost and treatment complexities, that are likely to limit its broad use in the near term. There have been few advances in therapies for SCD over the past two decades. Hydroxyurea (HU) induces HgbF which interrupts the polymerization of HgbS, and thereby has activity in decreasing the onset of vaso-occlusive crises and pathological sequelae of SCD. While HU is in wide use as a backbone therapy for SCD, it remains only partially effective, and is associated with toxicity, such as myelosuppression and teratogenicity. Patients receiving HU still experience hemolysis, anemia, and vaso-occlusive crises, suggesting a need for more effective therapies, either as a replacement or in combination with HU. Beyond HU, therapeutic intervention is largely supportive care, aimed at managing the symptoms of SCD. For instance, blood transfusions help with the anemia and other SCD complications by increasing the number of normal RBCs. However, repeated transfusions lead to iron overload and the need for chelation therapies to avoid consequent tissue damage. In addition to these approaches, analgesic medications are used to manage pain.


Given the current standard of care for SCD, there is a clear medical need for a noninvasive, disease-modifying therapy with appropriate safety and efficacy profiles.


SUMMARY

One aspect of the disclosure relates to methods of treating SCD comprising the administration of a therapeutically effective amount of a pyruvate kinase R (PKR) activator to a patient in need thereof diagnosed with SCD. Pyruvate kinase R (PKR) is the isoform of pyruvate kinase expressed in RBCs, and is a key enzyme in glycolysis. The invention is based in part on the discovery that the activation of PKR can target both sickling, by reducing deoxy-HgbS, and hemolysis. Targeting hemolysis may be achieved by improving RBC membrane integrity. One aspect of the disclosure is the recognition that activation of PKR can reduce 2,3-diphosphoglycerate (2,3-DPG), which leads to decreased deoxy-HgbS (and, therefore, sickling), as well as can increase ATP, which promotes membrane health and reduces hemolysis. Another aspect of the disclosure is the recognition that activation of PKR can reduce 2,3-diphosphoglycerate (2,3-DPG), which inhibits Hgb deoxygenation/increases oxygen affinity of HgbS and leads to decreased deoxy-HgbS (and, therefore, sickling), as well as can increase ATP, which promotes membrane health and reduces hemolysis. Accordingly, in one embodiment, PKR activation (e.g., by administration of a therapeutically effective amount of a PKR Activating Compound to a patient diagnosed with SCD) reduces RBC sickling via a reduction in levels of 2,3-diphosphoglycerate (2,3-DPG), which in turn reduces the polymerization of sickle Hgb (HgbS) into rigid aggregates that deform the cell. Furthermore, in some embodiments, PKR activation may contribute to overall RBC membrane integrity via increasing levels of adenosine triphosphate (ATP), which is predicted to reduce vaso-occlusive and hemolytic events which cause acute pain crises and anemia in SCD patients.


Preferably, a patient diagnosed with SCD is treated with a compound that is a PKR Activating Compound. The PKR activator can be a compound identified as a PKR Activating Compound or a composition identified as a PKR Activating Composition having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2, or a pharmaceutically acceptable salt and/or other solid form thereof. For example, the PKR Activating Compound can be the compound (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one (Compound 1):




embedded image



or a pharmaceutically acceptable salt thereof. Compound 1 is a selective, orally bioavailable PKR Activating Compound that decreases 2,3-DPG, increases ATP, and has anti-sickling effects in disease models with a wide therapeutic margin relative to preclinical toxicity.


PKR Activating Compounds can be readily identified as compounds of Formula I:




embedded image



or a pharmaceutically acceptable salt thereof, (e.g., Compound 1 and mixtures of Compound 1 with its stereoisomer) having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2.


In other embodiments, the PKR Activating Compound can be any of the compounds listed in FIG. 1, or a pharmaceutically acceptable salt thereof.


PKR Activating Compounds, such as 1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one, or a pharmaceutically acceptable salt thereof, are useful in pharmaceutical compositions for the treatment of patients diagnosed with SCD. PKR Activating Compounds, such as any of the compounds listed in FIG. 1, or a pharmaceutically acceptable salt thereof, are useful in pharmaceutical compositions for the treatment of patients diagnosed with SCD. The compositions comprising a compound of Formula I (e.g., Compound 1), or a pharmaceutically acceptable salt thereof, can be obtained by certain processes also provided herein. The compositions comprising any of the compounds listed in FIG. 1, or a pharmaceutically acceptable salt thereof, can be obtained by certain processes also provided herein.


The methods of treating SCD provided herein can offer greater protection against vaso-occlusive crises and hemolytic anemia, as compared to existing and emerging therapies. Therefore, use of a PKR Activating Compound, such as Compound 1, provides a novel and improved therapeutic approach either alone or in combination with drugs that act through alternative mechanisms, such as hydroxyurea (HU). In addition, use of a PKR Activating Compound, such as any of the compounds listed in FIG. 1, provides a novel and improved therapeutic approach either alone or in combination with drugs that act through alternative mechanisms, such as hydroxyurea (HU).





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a table of PKR Activating Compounds.



FIG. 2 is a schematic showing the relationship of PKR activation to the reduction of the clinical consequences of sickle cell disease (SCD).



FIG. 3 is a graph showing the oxyhemoglobin dissociation curve and modulating factors by plotting the relationship between hemoglobin saturation (percent) vs. partial pressure of oxygen (mmHg).



FIG. 4A is a chemical synthesis scheme for compounds of Formula I, including a synthesis of Compound 1 (separately provided in FIG. 4B).



FIG. 4B is a chemical synthesis scheme for Compound 1.



FIG. 4C is a general chemical synthesis of the compounds listed in FIG. 1.



FIG. 5 is a graph showing activation of recombinant PKR-R510Q with Compound 1, plotting the normalized rate vs. concentration of phosphoenolpyruvate (PEP) (Example 3).



FIG. 6 is a graph of data showing activation of recombinant PKR-R510Q by Compound 1 in the enzyme assay of Example 3.



FIG. 7 is a graph of data showing PKR activation in human red blood cells treated with Compound 1 (Example 4).



FIG. 8A (Study 1) and FIG. 8B (Study 2) are each graphs showing the observed changes in 2,3-DPG levels in blood from mice following 7 days of once daily (QD) oral treatment with Compound 1 (Example 5).



FIG. 9 is a graph showing observed changes in 2,3-DPG levels in blood from mice following 7 days of once daily (QD) oral treatment with Compound 1 (Example 5, Study 2).



FIG. 10A (Study 1) and FIG. 10B (Study 2) are graphs of data measuring ATP concentrations in red blood cells of mice following 7 days of once daily (QD) oral treatment with Compound 1 (Example 5).



FIG. 11 is a graph of the blood 2,3-DPG levels measured over time in healthy volunteers who received a single dose of Compound 1 or placebo.



FIG. 12 is a graph of the blood 2,3-DPG levels measured 24 hours post-dose in healthy volunteers who received a single dose of Compound 1 or placebo.



FIG. 13 is a graph of the blood 2,3-DPG levels measured over time in healthy volunteers who received daily doses of Compound 1 or placebo for 14 days.



FIG. 14 is a graph of the blood 2,3-DPG levels measured on day 14 in healthy volunteers who received daily doses of Compound 1 or placebo for 14 days.



FIG. 15 is a graph of the blood ATP levels measured on day 14 in healthy volunteers who received daily doses of Compound 1 or placebo for 14 days.



FIG. 16 is a graph plotting the blood concentration of Compound 1 (ng/mL) measured in healthy volunteer (HV) patients on a first (left) axis and the concentration of 2,3-DPG (micrograms/mL) measured in these HV patients on a second (right) axis after administration of a single dose of Compound 1 (400 mg).





DETAILED DESCRIPTION

Methods of treating SCD preferably include administration of a therapeutically effective amount of a compound (e.g., Compound 1) that reduces HgbS polymerization, for example by increasing HgbS affinity for oxygen. Methods of treating SCD also preferably include administration of a therapeutically effective amount of a compound (e.g., any of the compounds listed in FIG. 1) that reduces HgbS polymerization, for example by increasing HgbS affinity for oxygen. Methods of lowering 2,3-DPG and/or increasing ATP levels in human RBCs comprise administering a PKR Activating Compound, such as Compound 1. Methods of lowering 2,3-DPG and/or increasing ATP levels in human RBCs also comprise administering a PKR Activating Compound, such as any of the compounds listed in FIG. 1. Together these effects are consistent with providing therapies to reduce HgbS sickling and to improve RBC membrane health, presenting a unique disease-modifying mechanism for treating SCD.


A PKR Activating Compound, such as Compound 1, is useful to promote activity in the glycolytic pathway. A PKR Activating Compound, such as any of the compounds listed in FIG. 1, also is useful to promote activity in the glycolytic pathway. As the enzyme that catalyzes the last step of glycolysis, PKR directly impacts the metabolic health and primary functions of RBCs. PKR Activating Compounds (e.g., Compound 1), are useful to decrease 2,3-DPG and increase ATP. PKR Activating Compounds (e.g., any of the compounds listed in FIG. 1), are useful to decrease 2,3-DPG and increase ATP. PKR Activating Compounds (e.g., Compound 1 or any of the compounds listed in FIG. 1, preferably Compound 1) are also useful to increase Hgb oxygen affinity in RBC. The disclosure is based in part on the discovery that PKR activation is a therapeutic modality for SCD, whereby HgbS polymerization and RBC sickling are reduced via decreased 2,3-DPG and increased ATP levels.


SCD is the most common inherited blood disorder and clinically manifests with potentially severe pathological conditions associated with substantial physical, emotional, and economic burden. For instance, acute vaso-occlusive pain crises can be debilitating and necessitate rapid medical response. Chronic hemolytic anemia causes fatigue and often necessitates blood transfusions and supportive care. Over time, impaired oxygen transport through microvasculature precipitates organ and tissue damage. While there are a number of options available for treating symptoms, overall disease management would benefit from therapies that target upstream processes to prevent vaso-occlusion and hemolysis.


The described clinical symptoms are largely due to perturbations in RBC membrane shape and function resulting from aggregation of HgbS molecules. Unlike normal Hgb, HgbS polymerizes when it is in the deoxygenated state and ultimately causes a deformed, rigid membrane that is unable to pass through small blood vessels, thereby blocking normal blood flow through microvasculature. The loss of membrane elasticity also increases hemolysis, reducing RBC longevity. Furthermore, decreased cellular ATP and oxidative damage contribute to a sickle RBC membrane that is stiffer and weaker than that of normal RBCs. The damaged membrane has a greater propensity for adhering to vasculature, leading to hemolysis, increased aggregation of sickled RBCs, and increased coagulation and inflammation associated with vaso-occlusive crises.


The underlying cause of sickling is the formation of rigid deoxy-HgbS aggregates that alter the cell shape and consequently impact cellular physiology and membrane elasticity. These aggregates are highly structured polymers of deoxygenated HgbS; the oxygenated form does not polymerize. Polymerization is promoted by a subtle shift in conformation from the oxygen-bound relaxed (R)-state to the unbound tense (T)-state. In the latter, certain residues within the (β-chain of HgbS are able to interact in a specific and repetitive manner, facilitating polymerization.


The concentration of deoxy-HgbS depends on several factors, but the predominant factor is the partial pressure of oxygen (PO2). Oxygen reversibly binds to the heme portions of the Hgb molecule. As oxygenated blood flows via capillaries to peripheral tissues and organs that are actively consuming oxygen, PO2 drops and Hgb releases oxygen. The binding of oxygen to Hgb is cooperative and the relationship to PO2 levels fits a sigmoidal curve (FIG. 3). This relationship can be impacted by temperature, pH, carbon dioxide, and the glycolytic intermediate 2,3-DPG. 2,3-DPG binds within the central cavity of the Hgb tetramer, causes allosteric changes, and reduces Hgb's affinity for oxygen. Therefore, therapeutic approaches that increase oxygen affinity (i.e., reduce deoxygenation) of HgbS would presumably decrease polymer formation, changes to the cell membrane, and clinical consequences associated with SCD.


One aspect of this disclosure is targeting PKR activation to reduce 2,3-DPG levels, based on PKR's role in controlling the rate of glycolysis in RBCs. A decrease in 2,3-DPG with PKR activation has been demonstrated in preclinical studies and in healthy volunteers and patients with pyruvate kinase deficiency. Additionally, PKR activation would be expected to increase ATP, and has been observed to do so in these same studies. Given the role of ATP in the maintenance of a healthy RBC membrane and protection from oxidative stress, elevating its levels is likely to have broad beneficial effects. Therefore, activation of PKR offers the potential for a 2,3-DPG effect (i.e., reduced cell membrane damage from HgbS polymerization) that is augmented by ATP support for membrane integrity. It is via these changes that a PKR activator is could positively impact physiological changes that lead to the clinical pathologies of SCD (FIG. 2). In another aspect, the disclosure relates to a method of improving the anemia and the complications associated with anemia in SCD patients (e.g., ≥12 years of age) with Hgb SS or Hgb SB0-thalassemia.


Compound 1 is a selective, orally bioavailable PKR activator that has been shown to decrease 2,3-DPG, increase ATP, and have anti-sickling effects in disease models with a wide therapeutic margin relative to preclinical toxicity.


Methods of treatment can comprise administering to a subject in need thereof a therapeutically effective amount of (i) a PKR Activating Compound (e.g., a compound disclosed herein), or a pharmaceutically acceptable salt thereof; or (ii) a PKR Activating Composition (e.g., a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier). The pharmaceutical composition may be orally administered in any orally acceptable dosage form. In some embodiments, to increase the lifetime of red blood cells, a compound, composition, or pharmaceutical composition described herein is added directly to whole blood or packed cells extracorporeally or provided to the subject (e.g., the patient) directly. A patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject is in need of activation of PKR, and if the subject is determined to be in need of activation of PKR, then administering to the subject a PKR Activating Compound in a pharmaceutically acceptable composition. A patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject is diagnosed with SCD, and if the subject is diagnosed with SCD, then administering to the subject a PKR Activating Compound in a pharmaceutically acceptable composition. For example, administration of a therapeutically effective amount of a PKR Activating Compound can include administration of a total of about 25 mg-1,500 mg of Compound 1 each day, in single or divided doses. In some embodiments, Compound 1 is administered to patients diagnosed with SCD in total once daily (QD) doses of 25 mg, 50 mg, 75 mg, 100 mg, 125 mg, 150 mg, and/or higher if tolerated (e.g., 250 mg, 300 mg, 500 mg, 600 mg, 1000 mg, and/or 1500 mg). In some embodiments, a human dose of 80 to 130 mg of Compound 1 is administered once daily (QD) to a patient in need thereof (e.g., a patient diagnosed with SCD). In some embodiments, a PKR Activating Compound is administered in an amount of 400 mg per day (e.g., 400 mg QD or 200 mg BID). In some embodiments, Compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 400 mg per day (e.g., 400 mg QD or 200 mg BID). In some embodiments, any of the compounds listed in FIG. 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 400 mg per day (e.g., 400 mg QD or 200 mg BID). In some embodiments, a PKR Activating Compound is administered in an amount of 700 mg per day (e.g., 700 mg QD or 350 mg BID). In some embodiments, Compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 700 mg per day (e.g., 700 mg QD or 350 mg BID). In some embodiments, any of the compounds listed in FIG. 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 700 mg per day (e.g., 700 mg QD or 350 mg BID). In some embodiments, a PKR Activating Compound is administered in an amount of 100 mg, 200 mg, 400 mg, 600 mg, 700 mg, 1100 mg, or 1500 mg per day, in single or divided doses. In some embodiments, Compound 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 100 mg, 200 mg, 400 mg, 600 mg, 700 mg, 1100 mg, or 1500 mg per day, in single or divided doses. In some embodiments, any of the compounds listed in FIG. 1 or a pharmaceutically acceptable salt thereof is administered in an amount of 100 mg, 200 mg, 400 mg, 600 mg, 700 mg, 1100 mg, or 1500 mg per day, in single or divided doses.


Methods of treating a patient diagnosed with SCD can include administering to the patient in need thereof a therapeutic compound targeting reduction of deoxy-HgbS, which may or may not directly improve RBC membrane integrity. Compound 1 has been shown to decrease 2,3-DPG and increase ATP, and reduced cell sickling has been demonstrated in disease models. Accordingly, in some embodiments, the methods of treatment can address not only sickling, but also hemolysis and anemia.


Methods of treating a patient diagnosed with sickle cell disease, and PKR Activating Compounds for use in such methods, can include administering to the patient the PKR Activating Compound (e.g., a composition comprising one or more compounds of Formula I, such as Compound 1 or a mixture of Compound 1 and Compound 2) in an amount sufficient to reduce 2,3-DPG levels in the patient's red blood cells. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by at least 30% after 24 hours, or greater (e.g., reducing 2,3-DPG levels in the patient's red blood cells by at least 40% after 24 hours). In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 30-50% after 24 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 40-50% after 24 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by at least 25% after 12 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 25-45% after 12 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by at least 15% after 6 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 15-30% after 6 hours. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by at least 40% on day 14 of treatment. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 40-60% on day 14 of treatment. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by at least 50% on day 14 of treatment. In some embodiments, the amount is sufficient to reduce 2,3-DPG levels by 50-60% on day 14 of treatment.


Methods of treating a patient diagnosed with sickle cell disease, and PKR Activating Compounds for use in such methods, can also include administering to the patient the PKR Activating Compound (e.g., a composition comprising one or more compounds of Formula I, such as Compound 1 or a mixture of Compound 1 and Compound 2) in a daily amount sufficient to increase the patient's ATP blood levels. In some embodiments, the amount is sufficient to increase ATP blood levels by at least 40% on day 14 of treatment, or greater (e.g., at least 50% on day 14 of treatment). In some embodiments, the amount is sufficient to increase ATP blood levels by 40-65% on day 14 of treatment. In some embodiments, the amount is sufficient to increase ATP blood levels by at least 50% on day 14 of treatment, or greater (e.g., at least 50% on day 14 of treatment). In some embodiments, the amount is sufficient to increase ATP blood levels by 50-65% on day 14 of treatment.


In some examples, a pharmaceutical composition comprising Compound 1 can be used in a method of treating a patient diagnosed with sickle cell disease, the method comprising administering to the patient 400 mg of Compound 1 once per day (QD)




embedded image



In some examples, a pharmaceutical composition comprising Compound 1 can be used in a method of treating a patient diagnosed with sickle cell disease, the method comprising administering to the patient 200 mg of Compound 1 twice per day (BID)




embedded image



In some embodiments, the present disclosure provides PKR Activating Compounds of Formula I:




embedded image



or a pharmaceutically acceptable salt thereof. In some embodiments, a PKR Activating Compound is 1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one.


The compound of Formula I is preferably Compound 1:




embedded image



or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of Formula I is (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one.


In some embodiments, the present disclosure provides a PKR Activating Compound that is any of the compounds listed in FIG. 1, or a pharmaceutically acceptable salt thereof.


The present disclosure also provides compositions (e.g. pharmaceutical compositions) comprising a compound of Formula I. In some embodiments, a provided composition containing a compound of Formula I comprises a mixture of Compound 1 and Compound 2:




embedded image



or a pharmaceutically acceptable salt thereof. The present disclosure also provides compositions (e.g. pharmaceutical compositions) comprising any of the compounds listed in FIG. 1, or a pharmaceutically acceptable salt thereof.


Pharmaceutical compositions comprising a PKR Activating Composition containing a compound of Formula (I) can be formulated for oral administration (e.g., as a capsule or tablet). For example, Compound 1 can be combined with suitable compendial excipients to form an oral unit dosage form, such as a capsule or tablet, containing a target dose of Compound 1. The drug product can be prepared by first manufacturing Compound 1 as an active pharmaceutical ingredient (API), followed by roller compaction/milling with intragranular excipients and blending with extra granular excipients. A Drug Product can contain the Compound 1 API and excipient components in Table 1 in a tablet in a desired dosage strength of Compound 1 (e.g., a 25 mg or 100 mg tablet formed from a Pharmaceutical Composition in Table 1). The blended material can be compressed to form tablets and then film coated.


The pharmaceutical composition preferably comprises about 30-70% by weight of (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one, and a pharmaceutically acceptable excipient in an oral dosage form.









TABLE 1







Exemplary Pharmaceutical Compositions of


Compound 1 for Oral Administration










% Formulation



Function
(weight)
Examplary Component





API
  30-70%
Compound 1


Filler
  15-40%
Microcrystalline Cellulose


Drybinder
  2-10%
Crospovidone Kollidon CL


Glidant
0.25-1.25%
Colloidal Silicon Dioxide


Lubricant
0.25-1.00%
Magnesium Stearate, Hyqual









In some embodiments, a provided composition containing a compound of Formula I comprises a mixture of (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one and (R)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrollo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one. In some embodiments, a provided composition containing a compound of Formula I is a mixture of Compound 1 and Compound 2 as part of a PKR Activating Composition. In some embodiments, a compound of Formula I is racemic. In some embodiments, a compound of Formula I consists of about 50% of Compound 1 and about 50% of Compound 2. In some embodiments, a compound of Formula I is not racemic. In some embodiments, a compound of Formula I does not consist of about 50% of Compound 1 and about 50% of Compound 2. In some embodiments, a compound of Formula I comprises about 99-95%, about 95-90%, about 90-80%, about 80-70%, or about 70-60% of Compound 1. In some embodiments, a compound of Formula I comprises about 99%, 98%, 95%, 90%, 80%, 70%, or 60% of Compound 1.


In some embodiments, a PKR Activating Composition comprises a mixture of Compound 1 and Compound 2. In some embodiments, a PKR Activating Composition comprises a mixture of Compound 1 and Compound 2, wherein the PKR Activating Composition comprises a therapeutically effective amount of Compound 1.


Compositions comprising a compound of Formula I can be prepared as shown in FIG. 4A and FIG. 4B. Compounds of Formula I can be obtained by the general chemical synthesis scheme of FIG. 4A. Compound 1 can be obtained by the chemical synthesis route of FIG. 4A or FIG. 4B. In brief, compounds of Formula I (FIG. 4A) and/or Compound 1 (FIG. 4B) can be obtained from a series of four reaction steps from commercially available starting materials. Commercially available 7-bromo-2H,3H-[1,4]dioxino[2,3-b]pyridine was treated with a mixture of n-butyl lithium and dibutylmagnesium followed by sulfuryl chloride to give sulfonyl chloride 3. Treatment of 3 with tert-butyl 1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole-2-carboxylate in the presence of triethylamine (TEA) afforded Boc-protected monosulfonamide 4. Compound 4 was then de-protected in the presence of trifluoroacetic acid (TFA) to give 5, the free base of the monosulfonamide. The last step to generate Compound 1 (FIG. 4B) or Compound 1 and Compound 2 (FIG. 4A) was an amide coupling of 5 and tropic acid in the presence of 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluoro-phosphate (HATU).


The compounds listed in FIG. 1 can be prepared as shown in FIG. 4C and as described in International Publication No. WO 2018/175474, published Sep. 27, 2018. Generally, the compounds listed in FIG. 1 can be prepared by acylation and sulfonylation of the secondary amine groups of hexahydropyrrolopyrrole 6. For example, sulfonylation of 6 with a suitable sulfonyl chloride 7 affords sulfonyl hexahydropyrrolopyrrole 8, which is then treated with a suitable carboxylic acid 9 in the presence of an amide coupling reagent (e.g., HATU) to afford compound 10 (Path 1). Alternatively, acylation of 6 with a suitable carboxylic acid 9 in the presence of an amide coupling reagent affords acyl hexahydropyrrolopyrrole 11, which is then treated with a suitable sulfonyl chloride 7 to afford compound 10 (Path 2). As a person of ordinary skill would understand, well-known protecting groups, functionalization reactions, and separation techniques can be used in conjunction with Paths 1 and 2 to obtain the specific compounds listed in FIG. 1.


Methods of treating SCD also include administration of a therapeutically effective amount of a bioactive compound (e.g., a small molecule, nucleic acid, or antibody or other therapy) that reduces HgbS polymerization, for example by increasing HgbS affinity for oxygen.


In other embodiments, the disclosure relates to each of the following numbered embodiments:

    • 1. A composition comprising a PKR Activating Compound of Formula I, or a pharmaceutically acceptable salt thereof:




embedded image




    • 2. The composition of embodiment 1, wherein the compound of Formula I is Compound 1, or a pharmaceutically acceptable salt thereof:







embedded image




    • 3. The composition of embodiment 2, wherein the composition comprises a mixture of Compound 1 and Compound 2, or a pharmaceutically acceptable salt thereof:







embedded image




    • 4. The composition of embodiment 1, comprising the compound: 1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one.

    • 5. The composition of any one of embodiments 1-4, formulated as an oral unit dosage form.

    • 6. A method of treating a patient diagnosed with a sickle cell disease (SCD), the method comprising administering to the patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one, or a pharmaceutically acceptable salt thereof.

    • 7. The method of embodiment 6, wherein the method comprises oral administration of the pharmaceutical composition comprising (S)-1-(5-((2,3-dihydro-[1,4]dioxino[2,3-b]pyridin-7-yl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2-phenylpropan-1-one, as the only PKR Activating Compound in the pharmaceutical composition.

    • 8. A method of treating a patient diagnosed with a sickle cell disease (SCD), the method comprising administering to the patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising Compound 1:







embedded image



or a pharmaceutically acceptable salt thereof.

    • 9. A composition comprising a compound of Formula I obtainable by a process comprising the step of converting compound 5 into a compound of Formula I in a reaction described as Step 4:




embedded image




    • 10. The composition of embodiment 9, wherein the process further comprises first obtaining the compound 5 from a compound 4 by a process comprising Step 3:







embedded image




    • 11. The composition of embodiment 10, wherein the process further comprises first obtaining the compound 4 from a compound 3 by a process comprising Step 2:







embedded image




    • 12. The composition of embodiment 11, wherein the process further comprises first obtaining the compound 3 from a process comprising Step 1:







embedded image




    • 13. A method of treating a patient diagnosed with sickle cell disease (SCD), the method comprising administering to the patient in need thereof a therapeutically effective amount of a PKR Activating Compound having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2.

    • 14. The method of embodiment 13, wherein the PKR Activating Compound is Compound 1.

    • 15. The method of any one of embodiments 13-14, wherein the PKR Activating Compound is orally administered to the patient in need thereof.

    • 16. The use of Compound 1:







embedded image



or a pharmaceutically acceptable salt thereof, for the treatment of patients diagnosed with sickle cell disease (SCD).

    • 17. The use of a PKR Activating Compound having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2, in the treatment of patients diagnosed with sickle cell disease.
    • 18. The method of any one of embodiments 6-8 or 13-15, comprising the administration of Compound 1 once per day.
    • 19. The method of any one of embodiments 6-8 or 13-15, comprising the administration of a total of 25 mg-1,500 mg of Compound 1 each day.
    • 20. The method of any one of embodiments 18-19, comprising the administration of a total of 25 mg-130 mg of Compound 1 each day.


In other embodiments, the disclosure relates to each of the following numbered embodiments:

    • 1. A method for reducing 2,3-diphosphoglycerate (2,3-DPG) levels in a patient's red blood cells, comprising administering to the patient a PKR Activating Compound in a therapeutically effective amount, wherein the PKR Activating Compound is a compound of Formula I:




embedded image



or a pharmaceutically acceptable salt thereof, having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2.

    • 2. The method of embodiment 1, wherein the PKR Activating Compound is Compound 1:




embedded image



or a pharmaceutically acceptable salt thereof.

    • 3. The method of embodiment 1, wherein the PKR Activating Compound is Compound 1:




embedded image




    • 4. The method of embodiment 3, wherein the PKR Activating Compound is administered in an amount of 25-1500 mg per day.

    • 5. The method of embodiment 3, wherein the PKR Activating Compound is administered once daily in an amount of 250 mg, 300 mg, 500 mg, 600 mg, 1000 mg, or 1500 mg per day.

    • 6. The method of embodiment 3, wherein the PKR Activating Compound is administered once daily in an amount of 100 mg per day.

    • 7. The method of embodiment 3, wherein the PKR Activating Compound is administered once daily in an amount of 600 mg per day.

    • 8. The method of embodiment 3, wherein the PKR Activating Compound is administered once per day.

    • 9. The method of embodiment 3, wherein the PKR Activating Compound is orally administered to the patient.

    • 10. The method of embodiment 3, wherein Compound 1 is the only PKR Activating Compound administered to the patient.





In other embodiments, the disclosure relates to each of the following numbered embodiments:

    • 1. A method for reducing 2,3-diphosphoglycerate (2,3-DPG) levels in a patient's red blood cells, comprising administering to the patient the PKR Activating Compound in an amount sufficient to reduce 2,3-DPG levels in the patient's red blood cells by at least 30% after 24 hours, wherein the PKR Activating Compound is a compound of Formula I:




embedded image



or a pharmaceutically acceptable salt thereof, having an AC50 value of less than 1 μM using the Luminescence Assay described in Example 2.

    • 2. The method of embodiment 1, wherein the PKR Activating Compound is Compound 1:




embedded image



or a pharmaceutically acceptable salt thereof.

    • 3. The method of embodiment 1, wherein the PKR Activating Compound is Compound 1:




embedded image




    • 4. The method of embodiment 1, wherein Compound 1 is the only PKR Activating Compound administered to the patient.

    • 5. The method of any one of embodiments 1-4, wherein the PKR Activating Compound is orally administered to the patient.

    • 6. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered once per day.

    • 7. The method of any one of embodiments 1-6, wherein the PKR Activating Compound is administered in an amount sufficient to reduce 2,3-DPG levels in the patient's red blood cells by at least 40% after 24 hours.

    • 8. The method of any one of embodiments 1-7, wherein the PKR Activating Compound is administered in a daily amount sufficient to increase the patient's ATP blood levels by at least 40% on day 14 of treatment.

    • 9. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered in an amount of 100 mg, 200 mg, 400 mg, 600 mg, 700 mg, 1100 mg, or 1500 mg per day.

    • 10. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered in an amount of 200 mg per day.

    • 11. The method of embodiment 10, wherein the PKR Activating Compound is administered in an amount of 200 mg per day once per day (QD).

    • 12. The method of embodiment 10, wherein the PKR Activating Compound is administered in an amount of 100 mg per day twice per day (BID).

    • 13. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered in an amount of 400 mg per day.

    • 14. The method of embodiment 13, wherein the PKR Activating Compound is administered in an amount of 400 mg once per day (QD).

    • 15. The method of embodiment 13, wherein the PKR Activating Compound is administered in an amount of 200 mg twice per day (BID).

    • 16. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered in an amount of 600 mg per day.

    • 17. The method of embodiment 16, wherein the PKR Activating Compound is administered in an amount of 300 mg twice per day (BID).

    • 18. The method of any one of embodiments 1-5, wherein the PKR Activating Compound is administered in an amount of 700 mg per day.

    • 19. The method of embodiment 18, wherein the PKR Activating Compound is administered in an amount of 700 mg once per day (QD).

    • 30. The method of embodiment 18, wherein the PKR Activating Compound is administered in an amount of 350 mg twice per day (BID).





The present disclosure enables one of skill in the relevant art to make and use the inventions provided herein in accordance with multiple and varied embodiments. Various alterations, modifications, and improvements of the present disclosure that readily occur to those skilled in the art, including certain alterations, modifications, substitutions, and improvements are also part of this disclosure. Accordingly, the foregoing description and drawings are by way of example to illustrate the discoveries provided herein.


EXAMPLES

As the enzyme that catalyzes the last step of glycolysis, PKR underlies reactions that directly impact the metabolic health and primary functions of RBCs. The following Examples demonstrate how PKR activation by Compound 1 impacts RBCs. The primary effect of Compound 1 on RBCs is a decrease in 2,3-DPG that is proposed to reduce Hgb sickling and its consequences on RBCs and oxygen delivery to tissues. Compound 1 also increases ATP, which may provide metabolic resources to support cell membrane integrity and protect against loss of deformability and increased levels of hemolysis in SCD. With the combination of effects Compound 1 has on RBCs, it is likely to reduce the clinical sequelae of sickle Hgb and provide therapeutic benefits for patients with SCD.


The PKR Activating Compound designated Compound 1 was prepared as described in Example 1, and tested for PKR activating activity in the biochemical assay of Example 2.


The biological enzymatic activity of PKR (i.e., formation of ATP and/or pyruvate) was evaluated in enzyme and cell assays with Compound 1, as described in Example 3 and Example 4, respectively. Results from enzyme assays show that Compound 1 is an activator of recombinant wt-PKR and mutant PKR, (e.g., R510Q), which is one of the most prevalent PKR mutations in North America. PKR exists in both a dimeric and tetrameric state, but functions most efficiently as a tetramer. Compound 1 is an allosteric activator of PKR and is shown to stabilize the tetrameric form of PKR, thereby lowering the Km (the Michaelis-Menten constant) for PEP.


In vivo testing in mice (Examples 5) demonstrated PKR activation in wt mice, and provided an evaluation of effects on RBCs and Hgb in a murine model of SCD. Compound 1 was well tolerated up to the highest dose tested, and exposures increased in a dose-proportional manner. Levels of 2,3-DPG were reduced by >30% for doses 120 mg/kg Compound 1 (AUC from 0 to 24 hours (AUC0-24>5200 hr·ng/mL) and levels of ATP were increased by >40% for ≥60 mg/kg Compound 1 (AUC0-24>4000 hr·ng/mL).


In some embodiments, a daily dose of between 100 mg to 1500 mg of a PKR Activating Compound is administered to humans. In some embodiments, a daily dose of between 100 mg to 1500 mg of Compound 1 is administered to humans. In some embodiments, a daily dose of between 100 mg to 1500 mg of any of the compounds listed in FIG. 1 is administered to humans. In particular, a total daily dose of 100 mg-600 mg of a PKR Activating Compound can be administered to humans (including, e.g., a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg, per day, in single or divided doses). In particular, a total daily dose of 100 mg-600 mg of Compound 1 can be administered to humans (including, e.g., a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg, per day, in single or divided doses). In particular, a total daily dose of 100 mg-600 mg of any of the compounds listed in FIG. 1 can be administered to humans (including, e.g., a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg, per day, in single or divided doses). In some embodiments, a daily dose of 400 mg (e.g., 400 mg QD or 200 mg BID) of a PKR Activating Compound is administered to humans. In some embodiments, a daily dose of 400 mg (e.g., 400 mg QD or 200 mg BID) of Compound 1, or a pharmaceutically acceptable salt thereof, is administered to humans. In some embodiments, a daily dose of 400 mg (e.g., 400 mg QD or 200 mg BID) of any of the compounds listed in FIG. 1 is administered to humans.


Example 1: Synthesis of Compounds of Formula I

The PKR Activating Compound 1 was obtained by the method described herein and the reaction scheme shown in FIG. 4A and/or FIG. 4B. Compound 1 has a molecular weight of 457.50 Da.


Step 1. 2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl chloride (3)

Into a 100 mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of n-BuLi in hexane (2.5 M, 2 mL, 5.0 mmol, 0.54 equiv) and a solution of n-Bu2Mg in heptanes (1.0 M, 4.8 mL, 4.8 mmol, 0.53 equiv). The resulting solution was stirred for 10 min at RT (20° C.). This was followed by the dropwise addition of a solution of 7-bromo-2H,3H-[1,4]dioxino[2,3-b]pyridine (2 g, 9.26 mmol, 1.00 equiv) in tetrahydrofuran (16 mL) with stirring at −10° C. in 10 min. The resulting mixture was stirred for 1 h at −10° C. The reaction mixture was slowly added to a solution of sulfuryl chloride (16 mL) at −10° C. The resulting mixture was stirred for 0.5 h at −10° C. The reaction was then quenched by the careful addition of 30 mL of saturated ammonium chloride solution at 0° C. The resulting mixture was extracted with 3×50 mL of dichloromethane. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with ethyl acetate/petroleum ether (1:3). This provided 1.3 g (60%) of 2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl chloride as a white solid. LCMS m/z: calculated for C7H6ClNO4S: 235.64; found: 236 [M+H]+.


Step 2. tert-Butyl 5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole-2-carboxylate(4)

Into a 100-mL round-bottom flask was placed 2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl chloride (1.3 g, 5.52 mmol, 1.00 equiv), tert-butyl 1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole-2-carboxylate (1.16 g, 5.52 mmol), dichloromethane (40 mL), and triethylamine (1.39 g, 13.74 mmol, 2.49 equiv). The solution was stirred for 2 h at 20° C., then diluted with 40 mL of water. The resulting mixture was extracted with 3×30 mL of dichloromethane. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with dichloromethane/methanol (10:1). This provided 1.2 g (53%) of tert-butyl 5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole-2-carboxylate as a yellow solid. LCMS m/z: calculated for C18H23N3O6S: 409.46; found: 410 [M+H]+.


Step 3. 2-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole (5)

Into a 100-mL round-bottom flask was placed tert-butyl 5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole-2-carboxylate (1.2 g, 2.93 mmol, 1.00 equiv), dichloromethane (30 mL), and trifluoroacetic acid (6 mL). The solution was stirred for 1 h at 20° C. The resulting mixture was concentrated under vacuum. The residue was dissolved in 10 mL of methanol and the pH was adjusted to 8 with sodium bicarbonate (2 mol/L). The resulting solution was extracted with 3×10 mL of dichloromethane. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by silica gel column chromatography, eluting with dichloromethane/methanol (10:1). This provided 650 mg (72%) of 2-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole as a yellow solid. LCMS m/z: calculated for C13H15N3O4S: 309.34; found: 310 [M+H]+.


Step 4. (S)-1-(5-[2H, 3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one (1) and (R)-1-(5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one (2)

Into a 100 mL round-bottom flask was placed 2-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrole (150 mg, 0.48 mmol, 1.00 equiv), 3-hydroxy-2-phenylpropanoic acid (97 mg, 0.58 mmol, 1.20 equiv), dichloromethane (10 mL), HATU (369 mg, 0.97 mmol, 2.00 equiv) and DIEA (188 mg, 1.46 mmol, 3.00 equiv). The resulting solution was stirred overnight at 20° C. The reaction mixture was diluted with 20 mL of water and was then extracted with 3×20 mL of dichloromethane. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by prep-TLC eluted with dichloromethane/methanol (20:1) and further purified by prep-HPLC (Column:)(Bridge C18 OBD Prep Column, 100 Å, 5 μm, 19 mm×250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: MeCN; Gradient: 15% B to 45% B over 8 min; Flow rate: 20 mL/min; UV Detector: 254 nm). The two enantiomers were separated by prep-Chiral HPLC (Column, Daicel CHIRALPAK® IF, 2.0 cm×25 cm, 5 μm; mobile phase A: DCM, phase B: MeOH (hold 60% MeOH over 15 min); Flow rate: 16 mL/min; Detector, UV 254 & 220 nm). This resulted in peak 1 (2, Rt: 8.47 min) 9.0 mg (4%) of (R)-1-(5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one as a yellow solid; and peak 2 (1, Rt: 11.83 min) 10.6 mg (5%) of (S)-1-(5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one as a yellow solid.


(1): 1H NMR (400 MHz, DMSO-d6) δ 8.13 (d, J=2.0 Hz, 1H), 7.61 (d, J=2.0 Hz, 1H), 7.31-7.20 (m, 5H), 4.75 (t, J=5.2 Hz, 1H), 4.50-4.47 (m, 2H), 4.40-4.36 (m, 1H), 4.32-4.29 (m, 2H), 4.11-3.87 (m, 8H), 3.80-3.77 (m, 1H), 3.44-3.41 (m, 1H). LC-MS (ESI) m/z: calculated for C22H23N3O6S: 457.13; found: 458.0 [M+H]+.


(2): 1H NMR (400 MHz, DMSO-d6) δ 8.13 (d, J=2.0 Hz, 1H), 7.60 (d, J=2.0 Hz, 1H), 7.31-7.18 (m, 5H), 4.75 (t, J=5.2 Hz, 1H), 4.52-4.45 (m, 2H), 4.40-4.36 (m, 1H), 4.34-4.26 (m, 2H), 4.11-3.87 (m, 8H), 3.80-3.78 (m, 1H), 3.44-3.43 (m, 1H). LC-MS (ESI) m/z: calculated for C22H23N3O6S: 457.13; found: 458.0 [M+H]+.


Step 5. (S)-1-(5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one (1)

Alternatively, Compound 1 can be synthesized using the procedure described here as Step 5. A solution of 7-((3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1/H)-yl)sulfonyl)-2,3-dihydro-[1,4]dioxino[2,3-b]pyridine (130.9 mg, 0.423 mmol) in DMF (2.5 ml) was cooled on an ice bath, then treated with (S)-3-hydroxy-2-phenylpropanoic acid (84.8 mg, 0.510 mmol), HATU (195.5 mg, 0.514 mmol), and DIEA (0.30 mL, 1.718 mmol) and stirred at ambient temperature overnight. The solution was diluted with EtOAc (20 mL), washed sequentially with water (20 mL) and brine (2×20 mL), dried (MgSO4), filtered, treated with silica gel, and evaporated under reduced pressure. The material was chromatographed by Biotage MPLC (10 g silica gel column, 0 to 5% MeOH in DCM) to provide a white, slightly sticky solid. The sample was readsorbed onto silica gel and chromatographed (10 g silica gel column, 0 to 100% EtOAc in hexanes) to provide (2S)-1-(5-[2H,3H-[1,4]dioxino[2,3-b]pyridine-7-sulfonyl]-1H,2H,3H,4H,5H,6H-pyrrolo[3,4-c]pyrrol-2-yl)-3-hydroxy-2-phenylpropan-1-one (106.5 mg, 0.233 mmol, 55% yield) as a white solid.


Example 2: Biochemical Assay for Identification of PKR Activating Activity

PKR Activating Compounds can be identified with the biochemical Luminescence Assay of Example 2. The PKR activating activity of a series of chemical compounds was evaluated using the Luminescence Assay below, including compounds designated Compound 1, Compound 2, and Compounds 6, 7, and 8 below, and the compounds listed in FIG. 1.


For each tested compound, the ability to activate PKR was determined using the following Luminescence Assay. The effect of phosphorylation of adenosine-5′-diphosphate (ADP) by PKR is determined by the Kinase Glo Plus Assay (Promega) in the presence or absence of FBP (D-fructose-1,6-diphosphate; BOC Sciences, CAS: 81028-91-3) as follows. Unless otherwise indicated, all reagents are purchased from Sigma-Aldrich. All reagents are prepared in buffer containing 50 mM Tris-HCl, 100 mM KCl, 5 mM MgCl2, and 0.01% Triton X100, 0.03% BSA, and 1 mM DTT. Enzyme and PEP (phosphoenolpyruvate) are added at 2× to all wells of an assay-ready plate containing serial dilutions of test compounds or DMSO vehicle. Final enzyme concentrations for PKR(wt), PKR(R510Q), and PKR(G332S) are 0.8 nM, 0.8 nM, and 10 nM respectively. Final PEP concentration is 100 μM. The Enzyme/PEP mixture is incubated with compounds for 30 minutes at RT before the assay is initiated with the addition of 2×ADP and KinaseGloPlus. Final concentration of ADP is 100 μM. Final concentration of KinaseGloPlus is 12.5%. For assays containing FBP, that reagent is added at 30 μM upon reaction initiation. Reactions are allowed to progress for 45 minutes at RT until luminescence is recorded by the BMG PHERAstar FS Multilabel Reader. The compound is tested in triplicate at concentrations ranging from 42.5 μM to 2.2 nM in 0.83% DMSO. AC50 measurements were obtained by the standard four parameter fit algorithm of ActivityBase XE Runner (max, min, slope and AC50). The AC50 value for a compound is the concentration (μM) at which the activity along the four parameter logistic curve fit is halfway between minimum and maximum activity.


As set forth in Tables 2 and 3 below and in FIG. 1, AC50 values are defined as follows: ≤0.1 μM (+++); >0.1 μM and ≤1.0 μM (++); >1.0 μM and ≤40 μM (+); >40 μM (0).









TABLE 2







Luminescence Assay Data













AC50
AC50
AC50



Compound
(PKRG332S)
(PKRR510Q)
(WT)







1
++
+++
+++



2
+
+
+

















TABLE 3







Additional Luminescence Assay Data












AC50
AC50


Compound
Structure
(PKRG332S)
(PKRR510Q)





6


embedded image


++
+





7


embedded image


0
0





8


embedded image


0
0









Compounds and compositions described herein are activators of wild type PKR and certain PKR mutants having lower activities compared to the wild type. Such mutations in PKR can affect enzyme activity (catalytic efficiency), regulatory properties, and/or thermostability of the enzyme. One example of a PKR mutation is G332S. Another example of a PKR mutation is R510Q.


Example 3: Enzyme Assays of a PKR Activating Compound

The effect of 2 μM Compound 1 on maximum velocity (Vmax) and PEP Km (Michaelis-Menten constant, i.e., the concentration of PEP at which v=½ vmax) was evaluated for wt-PKR and PKR-R510Q. Tests were conducted in the presence and absence of fructose-1,6-bisphosphate (FBP), a known allosteric activator of PKR. Assessments were made up to 60 min at RT, and Vmax and PEP Km were calculated. The effect of Compound 1 on Vmax ranged from no effect to a modest increase (see FIG. 5 for a representative curve). Compound 1 consistently reduced the PEP Km, typically by ˜2 fold, for wt-PKR and PKR-R510Q in the presence or absence of FBP (Table 4), demonstrating that Compound 1 can enhance the rate of PKR at physiological concentrations of PEP.









TABLE 4







Effect of Compound 1 on PKR Enzyme Kinetic Parameters












No FBP
30 μM FBP













Kinetic

2 μM

2 μM


Enzyme
Parametera
DMSO
Compound 1
DMSO
Compound 1





WT-
Vmax
1.00
1.14
1.19
1.16


PKR
PEP Km
4.84
2.44
1.98
1.00


PKR
Vmax
1.54
1.56
1.00
1.29


R510Q
PEP Km
6.20
1.70
2.01
1.00






aAll values in Table 4 are normalized to 1.00, relative to the other values in the same row







Activation of wt-PKR and PKR-R510Q by different concentrations of Compound 1 was evaluated for PEP concentrations at or below Km. Compound 1 increased the rate of ATP formation, with AC50 values ranging from <0.05 to <0.10 μM and a range of <2.0 to <3.0 maximum-fold activation (i.e., <200% to <300%) (Table 5). Representative data from PKR-R510Q showed that the effect was concentration dependent (FIG. 6).









TABLE 5







Activation of PKR Wild and Mutant Types by Compound 1












Maximum-fold




PK Enzyme
Activation
AC50 (μM)







WT-PKR
<2.0
<0.05



PKR R510Q
<3.0
<0.10










Example 4: Cell Assays of a PKR Activating Compound

The activation of wt-PKR by Compound 1 in mature human erythrocytes ex vivo was evaluated in purified RBCs purchased from Research Blood Components. Cells treated with Compound 1 for 3 hr in glucose-containing media were washed, lysed, and assayed using a Biovision Pyruvate Kinase Assay (K709-100). The assay was repeated multiple times to account for donor-to-donor variability and the relatively narrow dynamic range. Mean maximum activation increase (Max-Min) was <100% and mean 50% effective concentration (EC50) was <125 nM (Table 6). wt-PKR was activated in a concentration-dependent manner (FIG. 7).









TABLE 6







Wild Type PKR Activation in


Human Red Blood Cells Treated with Compound 1









Replicate
Max − Min (%)
EC50 (nM)












1
<125
<250


2
<150
<150


3
<100
<50


4
<50
<50


Mean
<100
<125









Mouse RBCs were isolated fresh from whole blood using a Ficoll gradient and assayed with methods similar to those used in the human RBCs assays. Maximum activation increase, and EC50 values were comparable to the effects in human RBCs (Table 7).









TABLE 7







Effect of Compound 1 on PKR Activation


in Mouse Red Blood Cells









Replicate
Max − Min (%)
EC50 (nM)












1
<50
<125


2
<100
<125


Mean
<100
<125









Example 5: Pharmacokinetic/Pharmacodynamic Studies of Compound 1 in Wild Type Mice

Two pharmacokinetic (PK)/phamacodynamic (PD) studies were conducted in Balb/c mice that were administered Compound 1 once daily by oral gavage (formulated in 10% Cremophor EL/10% PG/80% DI water) for 7 days (QD×7) at doses of 0 (vehicle), 3.75, 7.5, 15, 60 mg/kg (Study 1); 0 (vehicle), 7.5, 15, 30, 60, 120, or 240 mg/kg (Study 2). On the 7th day, whole blood was collected 24 hours after dosing and snap frozen. Samples were later thawed and analyzed by LC/MS for 2,3-DPG and ATP levels. In both studies, Compound 1 was well tolerated. No adverse clinical signs were observed and there were no differences in body weight change compared with the vehicle group.


The levels of 2,3-DPG decreased with Compound 1 treatment (FIGS. 8A and 8B (Studies 1 and 2) and FIG. 9 (Study 2)). In general, reductions were >20% at ≥15 mg/kg Compound 1, and >30% for 120 and 240 mg/kg Compound 1. Together, the results from the highest doses provide in vivo evidence that 2,3-DPG decreases with PKR activation.


Evaluation of ATP levels in these studies showed that treatment with Compound 1 increased levels of ATP. In Study 1, ATP increased 21% and 79% with 30 and 60 mg/kg Compound 1, respectively, compared to vehicle, and in Study 2, ATP levels increased with exposure with doses up to 120 mg/kg Compound 1 with a maximum increase of ˜110% compared to vehicle (FIG. 10A and FIG. 10B). At the highest dose, 240 mg/kg Compound 1, ATP levels increased by 45%. Levels of ATP correlated with Compound 1 exposure in a manner similar across both studies.


Example 6: A SAD/MAD Study to Assess the Safety, Pharmacokinetics, and Pharmacodynamics of Compound 1 in Healthy Volunteers and Sickle Cell Disease Patients

Compound 1 will be evaluated in a randomized, placebo-controlled, double blind, single ascending and multiple ascending dose study to assess the safety, pharmacokinetics, and pharmacodynamics of Compound 1 in healthy volunteers and sickle cell disease patients. The use of Compound 1 is disclosed herein for treatment of sickle cell disease in humans.


Compound 1 is an oral small-molecule agonist of pyruvate kinase red blood cell isozyme (PKR) being developed for the treatment of hemolytic anemias. This human clinical trial study will characterize the safety, tolerability and the pharmacokinetics/pharmacodynamics (PK/PD) of a single ascending dose and multiple ascending doses of Compound 1 in the context of phase 1 studies in healthy volunteers and sickle cell disease patients. The effects of food on the absorption of Compound 1 will also be evaluated, in healthy volunteers.


The objectives of the study include the following:

    • 1. To evaluate the safety and tolerability of a single ascending dose and multiple ascending doses of Compound 1 in healthy volunteers and sickle cell disease (SCD) patients.
    • 2. To characterize the pharmacokinetics (PK) of Compound 1.
    • 3. To evaluate the levels of 2,3-diphosphoglycerate (DPG) and adenosine triphosphate (ATP) in the red blood cells (RBCs) of healthy volunteers and SCD patients after single and multiple doses of Compound 1.
    • 4. To evaluate the relationship between Compound 1 plasma concentration and potential effects on the QT interval in healthy volunteers.
    • 5. To evaluate the effect of single ascending doses of Compound 1 on other electrocardiogram (ECG) parameters (heart rate, PR and QRS interval and T-wave morphology) in healthy volunteers.
    • 6. To explore food effects on the PK of Compound 1 in healthy volunteers.
    • 7. To explore the association of Compound 1 exposure and response variables (such as safety, pharmacodynamics (PD), hematologic parameters as appropriate).
    • 8. To explore effects of Compound 1 after single and multiple doses on RBC function.
    • 9. To explore effects of Compound 1 after multiple doses in SCD patients on RBC metabolism, inflammation and coagulation.


This is a first-in-human (FIH), Phase 1 study of Compound 1 that will characterize the safety, PK, and PD of Compound 1 after a single dose and after repeated dosing first in healthy adult volunteers and then in adolescents or adults with sickle cell disease. The study arms and assigned interventions to be employed in the study are summarized in Table 8. Initially, a dose range of Compound 1 in single ascending dose (SAD) escalation cohorts will be explored in healthy subjects. Enrollment of healthy subjects into 2-week multiple ascending dose (MAD) escalation cohorts will be initiated once the safety and PK from at least two SAD cohorts is available to inform the doses for the 2-week MAD portion of the study. The MAD cohorts will then run in parallel to the single dose cohorts. A single dose cohort is planned to understand food effects (FE) on the PK of Compound 1. After the SAD and FE studies in healthy subjects are completed, the safety, PK and PD of a single dose of Compound 1 that was found to be safe in healthy subjects will then be evaluated in sickle cell disease (SCD) subjects. Multiple dose studies in SCD subjects will then be initiated upon completion of MAD studies in healthy volunteers. Compound 1 will be administered in 25 mg and 100 mg tablets delivered orally.










TABLE 8





Arms
Assigned Interventions







Experimental: Single ascending dose
Drug: Compound 1/Placebo


cohorts in healthy subjects
Healthy volunteer subjects


Healthy volunteer subject cohorts
will receive Compound 1/


randomized 6:2 receiving a single dose of
placebo and be monitored


Compound 1 or placebo. The first cohort
for side effects while


will receive 200 mg of Compound 1 or
undergoing pharmacokinetic


placebo. Dose escalation will occur if
and pharmacodynamics


Compound 1 or placebo is tolerated. The
studies


maximum dose of Compound 1 or



placebo will be 1500 mg. Planned doses



for the SAD cohorts are listed in Table 9.



Experimental: Multiple ascending dose
Drug: Compound 1/


cohorts in healthy subjects
Placebo


Healthy volunteer subject cohorts
Healthy volunteer


randomized 9:3 to receive Compound 1 or
subjects will receive


placebo for 14 days continuous dosing.
Compound 1/placebo and


The first cohort will receive 100 mg of
be monitored for side


Compound 1 or placebo daily × 14 days.
effects while undergoing


Alternatively, the first cohort will receive
pharmacokinetics and


200 mg (e.g., 100 mg BID or 200 mg QD)
pharmacodynamic studies


of Compound 1 or placebo daily × 14



days. The maximum dose of Compound



1/placebo will be 600 mg Compound



1/placebo daily for 14 days. Planned



doses for the MAD cohorts are listed in



Table 10.



Experimental: Food Effect Cohort in
Drug: Compound 1


healthy subjects
Healthy subjects will


Healthy Volunteer subject cohort of 10
receive Compound 1 with


subjects who will receive a single dose of
or without food and


Compound 1 with food and without food.
undergo pharmacokinetic


Dose will be administered per the protocol
studies


defined dose. Healthy Volunteer subject



cohort of 10 subjects who will receive a



single dose of Compound 1 with food and



without food. Dose will be 500 mg of



Compound 1, but is subject to change



based on the pharmacokinetic profile of



Compound 1 observed in the initial SAD



cohorts and the safety profile of



Compound 1 observed in prior SAD and



MAD cohorts.



Experimental: Single ascending dose
Drug: Compound 1/


cohorts in SCD subjects
Placebo


Sickle cell disease subject cohort
SCD subjects


randomized 6:2 receiving a single dose of
will receive Compound


Compound 1 or placebo. The dose of
1/placebo and be


Compound 1/placebo administered will be
monitored for side


a dose that was found to be safe in healthy
effects while undergoing


subjects. The dose of Compound
pharmacokinetic and


1/placebo administered also will be a dose
pharmacodynamics


that was found to be
studies


pharmacodynamically active (e.g., results



in a reduction in 2,3-DPG) in healthy



subjects.



Experimental: Multiple ascending dose
Drug: Compound 1/


cohorts in SCD subjects
Placebo


Sickle cell disease subject cohorts
SCD subjects


randomized 9:3 to receive Compound 1 or
will receive Compound


placebo for 14 days continuous dosing.
1/placebo and be


The dose of Compound 1/placebo
monitored for side


administered will be a dose less than
effects while undergoing


maximum tolerable dose evaluated in
pharmacokinetic and


MAD healthy volunteers. The dose of
pharmacodynamics


Compound 1/placebo also will be a dose
studies


that was found to be



pharmacodynamically active (e.g., results



in a reduction in RBC 2,3-DPG and



increase in RBC ATP) in MAD healthy



volunteers.




















TABLE 9







Dose Level/Cohort
Dose
Tablet Strength (#/day)









SAD 1
 200 mg
100 mg (2/day)



SAD 2
 400 mg
100 mg (4/day)



SAD 3
 700 mg
100 mg (7/day)



SAD 4
1100 mg
 100 mg (11/day)



SAD 5
1500 mg
 100 mg (15/day)





















TABLE 10







Dose Level/Cohort
Dose
Tablet Strength (#/day)









MAD 1
100 mg
 100 mg (1/day) or





 25 mg (4/day)



MAD 2
200 mg
100 mg (2/day)



MAD 3
400 mg
100 mg (4/day)



MAD 4
600 mg
100 mg (6/day)










Outcome Measures

Primary Outcome Measures:






    • 1. Incidence, frequency, and severity of adverse events (AEs) per CTCAE v5.0 of a single ascending dose and multiple ascending doses of Compound 1 in adult healthy volunteers and SCD patients.
      • [Time Frame: Up to 3 weeks of monitoring]

    • 2. Maximum observed plasma concentration (Cmax)
      • [Time Frame: Up to 3 weeks of testing]

    • 3. Time to maximum observed plasma concentration (Tmax)
      • [Time Frame: Up to 3 weeks of testing]

    • 4. Area under the plasma concentration-time curve from time zero until the 24-hour time point (AUC0-24)
      • [Time Frame: Up to 3 weeks of testing]

    • 5. Area under the plasma concentration-time curve from time zero until last quantifiable time point (AUC0-last)
      • [Time Frame: Up to 3 weeks of testing]

    • 6. Area under the plasma concentration-time curve from time zero to infinity (AUC0-inf)
      • [Time Frame: Up to 3 weeks of testing]

    • 7. Terminal elimination half-life (t1/2)
      • [Time Frame: Up to 3 weeks of testing]

    • 8. Apparent clearance (CL/F)
      • [Time Frame: Up to 3 weeks of testing]

    • 9. Apparent volume of distribution (Vd/F)
      • [Time Frame: Up to 3 weeks of testing]

    • 10. Terminal disposition rate constant (Lz)
      • [Time Frame: Up to 3 weeks of testing]

    • 11. Renal clearance (CIR)
      • [Time Frame: Up to 3 weeks of testing]


        Secondary Outcome Measures:

    • 12. Change from baseline in the levels of 2,3-diphosphoglycerate (DPG) and adenosine triphosphate (ATP) in the red blood cells (RBCs) of healthy volunteers and SCD patients after single and multiple doses of Compound 1.
      • [Time Frame: Up to 3 weeks of testing]

    • 13. Model-based estimate of change from baseline QT interval corrected using Fridericia's correction formula (QTcF) and 90% confidence interval at the estimated Cmax after a single dose of Compound 1 in healthy volunteers.
      • [Time Frame: up to 7 days]

    • 14. Change from baseline heart rate after a single dose of Compound 1 in healthy volunteers
      • [Time Frame: up to 7 days]

    • 15. Change from baseline PR after a single dose of Compound 1 in healthy volunteers
      • [Time Frame: up to 7 days]

    • 16. Change from baseline QRS after a single dose of Compound 1 in healthy volunteers
      • [Time Frame: up to 7 days]

    • 17. Change from baseline T-wave morphology after a single dose of Compound 1 in healthy volunteers
      • [Time Frame: up to 7 days]


        Exploratory Outcome Measures:

    • 18. Effect of food on Cmax, AUC0-4/AUClast

    • 19. Effect of AUClast/AUC0-24, Cmax, minimum plasma concentration (Cmin), peak-to trough ratio, dose linearity, accumulation ratio on safety, PD, and hematologic parameters of interest, as assessed by exposure-response analyses

    • 20. Effect of chronic Compound 1 dosing on SCD RBC response to oxidative stress in SCD Patients (including evaluation of glutathione, glutathione peroxidase and superoxide dismutase levels)

    • 21. Effect of chronic Compound 1 dosing on measurable markers of inflammation in SCD Patients (C-reactive protein, ferritin, interleukin [IL]-1β, IL-6, IL-8, and tumor necrosis factor-α)

    • 22. Effects of chronic Compound 1 dosing on measurable markers of hypercoagulation in SCD patients (D-dimer, prothrombin 1.2, and thrombin-antithrombin [TAT] complexes)





Eligibility





    • Minimum age: 18 Years (healthy volunteers); 12 Years (SCD subjects)

    • Maximum age: 60 Years

    • Sex: All

    • Gender Based: No

    • Accepts Healthy Volunteers: Yes


      Inclusion Criteria:

    • Healthy volunteer: subjects must be between 18 and 60 years of age; SCD: subjects must be between 12 and 50 years of age

    • Subjects must have the ability to understand and sign written informed consent, which must be obtained prior to any study-related procedures being completed.

    • Subjects must be in general good health, based upon the results of medical history, a physical examination, vital signs, laboratory profile, and a 12-lead ECG.

    • Subjects must have a body mass index (BMI) within the range of 18 kg/m2 to 33 kg/m2 (inclusive) and a minimum body weight of 50 kg (healthy volunteer subjects) or 40 kg (SCD subjects)

    • For SCD subjects, sickle cell disease previously confirmed by hemoglobin electrophoresis or genotyping indicating one of the following hemoglobin genotypes: Hgb SS, Hgb Sβ+-thalassemia, Hgb Sr-thalassemia, or Hgb SC

    • All males and females of child bearing potential must agree to use medically accepted contraceptive regimen during study participation and up to 90 days after.

    • Subjects must be willing to abide by all study requirements and restrictions.


      Exclusion Criteria:

    • Evidence of clinically significant medical condition or other condition that might significantly interfere with the absorption, distribution, metabolism, or excretion of study drug, or place the subject at an unacceptable risk as a participant in this study

    • History of clinically significant cardiac diseases including condition disturbances Abnormal hematologic, renal and liver function studies

    • History of drug or alcohol abuse





Results (Healthy Volunteers)

Four healthy SAD cohorts were evaluated at doses of 200, 400, 700, and 1000 mg, and four healthy MAD cohorts received 200 to 600 mg total daily doses for 14 days at QD or BID dosing (100 mg BID, 200 mg BID, 300 mg BID, and 400 mg QD). In the food effect (FE) cohort, 10 healthy subjects received 200 mg of Compound 1 QD with and without food.


No serious adverse events (SAEs) or AEs leading to withdrawal were reported in the SAD and MAD cohorts of healthy volunteers. In PK assessments, Compound 1 was rapidly absorbed with a median Tmax of 1 hr postdose. Single dose exposure increased in greater than dose-proportional manner at doses ≥700 mg. In multiple-doses delivered BID or QD, linear PK was observed across all dose levels (100-300 mg BID, 400 mg QD), and exposure remained steady up to day 14, without cumulative effect. Compound 1 exposure under fed/fasted conditions was similar.


PD activity was demonstrated at all dose levels evaluated in Compound 1-treated subjects (Table 11). Table 11 reports the mean maximum percentage change in 2,3-DPG and ATP across all doses and timepoints in the SAD and MAD cohorts. As shown in Table 11, a mean decrease in 2,3-DPG, and a mean increase in ATP, relative to baseline, was observed in both the SAD and MAD cohorts. Within 24 hr of a single dose of Compound 1, a decrease in 2,3-DPG was observed. After 14 days of Compound 1 dosing these PD effects were maintained along with an increase in ATP over baseline. Accordingly, the mean maximum reduction in the concentration of 2,3-DPG was at least about 40% in patients receiving Compound 1 in the SAD study and at least about 50% in patients receiving Compound 1 in the MAD study.









TABLE 11







Summary of Mean Maximum Percent Change


in Key PD Measures from Baseline












SAD
MAD














Placebo
Compound 1
Placebo
Compound 1


PD Marker
Statistics
(N = 8)
(N = 24)
(N = 12
(N = 36)















2,3-DPG
Mean
−19.5
−46.8
−17.0
−56.3



(95% CI)
(−25.0, −14.0)
(−50.3, −43.2)
(−22.9, −11.1)
(−58.9, −53.7)



P-value

<0.0001

<0.0001


ATP
Mean
9.2
24.4
7.2
68.5



(95% CI)
 (0.5, 18.0)
(18.4, 30.3)
 (−0.3, 14.7)  
(63.6, 73.3)



P-value

0.0094

<0.0001









In the SAD cohorts, the subjects' blood 2,3-DPG levels were measured periodically after dosing by a qualified LC-MS/MS method for the quantitation of 2,3-DPG in blood. Decreased 2,3-DPG blood levels were observed 6 hours following a single dose of Compound 1 at all dose levels (earlier timepoints were not collected). Maximum decreases in 2,3-DPG levels generally occurred ˜24 hours after the first dose with the reduction sustained ˜48-72 hr postdose. Table 12 reports the median percentage change in 2,3-DPG blood levels, relative to baseline, measured over time in healthy volunteers after a single dose of Compound 1 (200 mg, 400 mg, 700 mg, or 1000 mg) or placebo. Accordingly, the median reduction in the concentration of 2,3-DPG, relative to baseline, was at least about 30% at all dose levels tested 24 hours after administration of the single dose.









TABLE 12







Median Percentage Change in 2,3-DPG Levels








Time After
Dose












Dose
Placebo
200 mg
400 mg
700 mg
1000 mg















0
0.0
0.0
0.0
0.0
0.0


6
−7.8
−18
−23
−29
−20


8
−7.6
−17
−29
−28
−31


12
−4.0
−25
−40
−41
−44


16
−6.0
−33
−35
−46
−50


24
−2.0
−31
−39
−49
−48


36
−6.9
−33
−38
−46
−47


48
−15
−29
−31
−48
−47


72
−6.9
−18
−30
−33
−21










FIG. 11 is a graph of the blood 2,3-DPG levels measured over time in healthy volunteers who received a single dose of Compound 1 (200 mg, 400 mg, 700 mg, or 1000 mg) or placebo. As shown in FIG. 11, healthy volunteers who received Compound 1 experienced a decrease in blood 2,3-DPG levels, relative subjects who received the placebo. FIG. 12 is a graph of the blood 2,3-DPG levels measured 24 hours post-dose in healthy volunteers who received a single dose of Compound 1 (200 mg, 400 mg, 700 mg, or 1000 mg) or placebo. As shown in FIG. 12, healthy volunteers who received Compound 1 experienced a decrease in blood 2,3-DPG levels at 24 hours post-dose, relative to subjects who received the placebo.


In the MAD cohorts, the subjects' blood 2,3-DPG levels were measured periodically after dosing by a qualified LC-MS/MS method for the quantitation of 2,3-DPG in blood. The maximum decrease in 2,3-DPG on Day 14 was 55% from baseline (median). 2,3-DPG levels reached a nadir and plateaued on Day 1 and had not returned to baseline levels 72 hours after the final dose on Day 14. Table 13 reports the median percentage change in 2,3-DPG blood levels, relative to baseline, measured over time after the first dose on days 1 and 14 in healthy volunteers who received daily doses of Compound 1 (100 mg BID, 200 mg BID, or 300 mg BID) or placebo for 14 days. Accordingly, the median reduction in the concentration of 2,3-DPG, relative to baseline, was at least about 25% at all dose levels tested 24 hours after administration of the first dose on day 1 and at least about 40% at all dose levels tested 24 hours after administration of the first dose on day 14.









TABLE 13







Median Percentage Change in 2,3-DPG Levels (Days 1 and 14)









Dose











Time After
100 mg BID
200 mg BID
300 mg BID
Placebo


First Daily
Day
Day
Day
Day















Dose
1
14
1
14
1
14
1
14


















0
0.0
−42.0
0.0
−48.2
0.0
−59.4
0.0
−7.6


6
−16.1
−44.3
−13.1
−48.5
−18.8
−53.0
−2.9
−10.9


8
−12.1
−44.7
−22.3
−44.3
−23.8
−54.2
−0.6
−1.6


12
−18.1
−43.6
−23.1
−42.2
−31.6
−55.3
−7.1
−1.6


16
−18.4
−43.9
−33.9
−42.9
−40.7
−52.4
−6.7
−5.3


24
−27.8
−44.1
−43.5
−44.3
−50.8
−52.1
1.1
−10.7


48

−34.7

−38.7

−44.5

−1.0


72

−20.2

−20.2

−32.9

−7.0










FIG. 13 is a graph of the blood 2,3-DPG levels measured over time in healthy volunteers who received daily doses of Compound 1 (100 mg BID, 200 mg BID, 300 mg BID, or 400 mg QD) or placebo for 14 days. As shown in FIG. 13, healthy volunteers who received Compound 1 experienced a decrease in blood 2,3-DPG levels, relative subjects who received the placebo. FIG. 14 is a graph of the blood 2,3-DPG levels measured on day 14 in healthy volunteers who received daily doses of Compound 1 (100 mg BID, 200 mg BID, 300 mg BID, or 400 mg QD) or placebo for 14 days. As shown in FIG. 14, healthy volunteers who received Compound 1 experienced a decrease in blood 2,3-DPG levels, relative to subjects who received the placebo.


In the MAD cohorts, the subjects' blood ATP levels were measured on day 14 by a qualified LC-MS/MS method for the quantitation of ATP in blood. ATP levels were elevated, relative to baseline, on day 14, and remained elevated 60 hours after the last dose. Table 14 reports the median percentage change in blood ATP levels, relative to baseline, measured over time after the first dose on day 14 in healthy volunteers who received daily doses of Compound 1 (100 mg BID, or 200 mg BID) or placebo for 14 days.









TABLE 14







Median Percentage Change in ATP Levels (Day 14)










Time After First
Dose












Daily Dose
100 mg BID
200 mg BID
Placebo
















0
41.5
55.3
−0.5



6
43.8
48.1
2.8



8
47.8
58.4
−4.1



12
45.4
56.2
2.3



16
44.8
57.0
−6.8



24
55.0
64.0
2.9



48
52.2
58.9
4.7



72
49.2
54.0
2.2











FIG. 15 is a graph of the blood ATP levels measured on day 14 in healthy volunteers who received daily doses of Compound 1 (100 mg BID, 200 mg BID, 300 mg BID, or 400 mg QD) or placebo for 14 days. As shown in FIG. 15, healthy volunteers who received Compound 1 experienced an increase in blood ATP levels, relative to subjects who received the placebo.



FIG. 16 is a graph plotting the blood concentration of Compound 1 (ng/mL) measured in healthy volunteer (HV) patients on a first (left) axis and the concentration of 2,3-DPG (micrograms/mL) measured in these HV patients on a second (right) axis after administration of a single dose of Compound 1 (400 mg). Solid symbols represent geometric means and Standard errors of the observed Compound 1 plasma and 2,3 DPG concentrations. As shown in the figure, the observed 2,3 DPG modulation does not track directly plasma pharmacokinetics (blood concentration of Compound 1) where the pharmacodynamic maximum (i.e., the minimum of the 2,3-DPG concentration, at time ˜24 h) occurred nearly 24 h after the pharmacokinetic maximum (i.e., maximum of the PK curve, at time ˜1-2 h). The observed pharmacodynamic response in HVs was durable, with a calculated PD half-life of ˜20 h, where 2,3-DPG depression was observed long after plasma levels were undetectable. Taken together, this suggests that identifying the pharmacologically active dose cannot be adequately performed using pharmacokinetic parameters (Cmax/Cmin/AUC) in isolation, but rather support an approach that includes integrating the temporal pharmacokinetic/pharmacodynamic relationship to provide the platform of evidence that QD dosing may be feasible in sickle cell disease patients.


Example 7: Analysis of ATP and 2,3 DPG in K2EDTA Whole Blood by LC-MS/MS

The following procedures are employed for the analysis of ATP and 2,3-DPG in human whole blood K2EDTA using a protein precipitation extraction procedure and analysis by LC-MS/MS.


This bioanalytical method applies to the parameters described below:















Assay Range
25,000-1,500,000 ng/mL


Extraction Volume
15.0 μL


Species/Matrix/Anticoagulant
Water as a surrogate for Human Whole



Blood K2EDTA


Extraction type
Protein Precipitation


Sample Storage
80° C.


Mass Spectrometer
API-5500


Acquisition software
Analyst/Aria System









The following precautions are followed:

    • 1. Standard and QC samples are prepared on ice and stored in plastic containers.
    • 2. Study samples and QC samples are thawed on ice.
    • 3. Extraction is performed on ice.


The following definitions and abbreviations are employed:


















CRB
Carryover remediation blanks



FT
Freeze-thaw



MPA
Mobile phase A



MPB
Mobile phase B



NA
Not applicable



NR
Needle rinse



RT
Retention time



SIP
Stability in progress



TBD
To be determined










The following chemicals, matrix, and reagents are used:














K2EDTA Human Whole Blood, BioreclamationIVT or equivalent


(Note: BioReclamationIVT and BioIVT are considered equivalent)


Acetonitrile (ACN), HPLC Grade or better


Ammonium Acetate (NH4OAc), HPLC grade or equivalent


Ammonium Hydroxide (NH4OH, 28-30%), ACS grade or better


Dimethylsulfoxide (DMSO), ACS grade or better


Formic Acid (FA), 88% ACS grade


Isopropanol (IPA), HPLC Grade or better


Methanol (MeOH), HPLC Grade or better


Water (H2O), Milli-Q or HPLC Grade


ATP-Analyte, Sponsor or supplier


ATP-IS-IS, Sponsor or supplier


2,3-DPG-Analyte, Sponsor or supplier


2,3-DPG-IS-IS, Sponsor or supplier









The following procedures are used for reagent preparation. Any applicable weights and volumes listed are nominal and may be proportionally adjusted as long as the targeted composition is achieved:

















Nominal Volumes




Final Solution
for Solution
Storage


Solution
Composition
Preparation
Conditions







Mobile Phase A
10 mM Ammoniumn
Weigh
Ambient


(MPA)
Acetate in water pH
approximately
Temperature



8.5
770.8 mg of





Ammonium





Acetate; add to a





bottle with 1000 mL





of water.





Adjust pH to 8.3-8.7





using Ammonium





Hydroxide.



Mobile Phase B
5:95 MPA:ACN
Add 50.0 mL of
Ambient


(MPB)

MPA to 950 mL of
Temperature




CAN. Mix.



Needle Rinse 1
25:25:25:25:0.1
Add 500 mL of
Ambient


(NR1)
(v:v:v:v:v)
MeOH, 500 mL of
Temperature



MeOH:ACN:H2O:
ACN, 500 mL of




IPA:NH4OH
H2O, 500 mL of





IPA, and 2 mL of





NH4OH. Mix.



Needle Rings 2
90:10:0.1 (v:v:v)
Add 2 mL of FA to
Ambient


(NR2)
H20:MeOH:FA
200 mL of MeOH
Temperature




and 1800 mL of





H20. Mix.









Calibration standards are prepared using water as the matrix according to the table presented below. The indicated standard is prepared by diluting the indicated spiking volume of stock solution with the indicated matrix volume.




















Stock
Spiking
Matrix
Final
Final


Calibration
Stock
Conc.
Vol.
Vol.
Vol.
Conc.


Standard
Solution
(ng/mL)
(mL)
(mL)
(mL)
(ng/mL)





















STD-6
ATP
60,000,000
0.0100
0.380
0.400
1,500,000



Stock








2,3-DPG
60,000,000
0.0100






Stock







STD-5
STD-6
1,500,000
0.100 
0.200
0.300
500,000


STD-4
STD-6
1,500,000
0.0500
0.325
0.375
200,000


STD-3
STD-6
1,500,000
0.0250
0.350
0.375
100,000


STD-2
STD-5
500,000
0.0500
0.450
0.500
50,000


STD-1
STD-5
500,000
0.0250
0.475
0.500
25,000


Cond.
STD-5
500,000
0.0250
0.975
1.00 
12,500









Quality control standards are prepared using water as the matrix according to the table presented below. The indicated quality control standard is prepared by diluting the indicated spiking volume of stock solution with the indicated matrix volume.


















Quality

Stock
Spiking
Matrix
Final
Final


Control
Stock
Conc.
Vol.
Vol.
Vol.
Conc.


Standard
Solution
(ng/mL)
(mL)
(mL)
(mL)
(ng/mL)





















QC-High
ATP
60,000,000
0.160
7.68
8.00
1,200,000



Stock








2,3-DPG
60,000,000
0.160






Stock







QC-Mid
QC-High
1,200,000
1.50 
4.50
6.00
300,000


QC-Low
QC-Mid
300,000
1.50 
4.50
6.00
75,000









An internal standard spiking solution is prepared with a final concentration of 12,500 ng/mL ATP and 2,3-DPG by diluting stock solutions of ATP and 2,3-DPG at concentrations of 1,000,000 ng/mL with water. 0.200 mL each of the ATP and 2,3-DPG stock solutions are diluted with 15.6 mL of water to produce a final volume of 16.0 mL at a final concentration of 12,500 ng/mL of ATP and 2,3-DPG.


The following procedures are used for sample extraction prior to analysis via LC-MS/MS. 15.0 μL of the calibration standards, quality controls, matrix blanks, and samples are aliquoted into a 96-well plate. 50.0 μL of the internal standard spiking solution is added to all samples on the plate, with the exception of the matrix blank samples; 50.0 μL of water is added to the matrix blank samples. Subsequently, 150 μL of water is added to all samples on the plate. The plate is then covered and agitated by vortex at high speed for ten minutes, after which 750 μL of methanol is added to all samples on the plate. The plate is covered and agitated by vortex for approximately 1 minute. The plate is then centrifuged at approximately 3500 RPM at approximately 4° C. for five minutes. After centrifugation, a liquid handler is used to transfer 50 μL of each sample to a new 96-well plate, and 200 μL of acetonitrile is added to all samples on the plate. The newly prepared plate is covered and agitated by vortex for approximately 1 minute. The plate is then centrifuged at approximately 3500 RPM at approximately 4° C. for 2 minutes.


The following LC parameters and gradient conditions are used for analysis of the extracted samples:












LC Parameters

















Analytical Column
Vendor:
SeQuant



Description:
ZIC-pHILIC



Dimensions:
50 mm × 2.1 mm



Column Heater Temperature:
40° C.


Plate Rack
Position:
Cold Stack



Cold Stack Set Point:
 5° C.


Mobile Phase
Mobile Phase A
10 mM Ammoniumn



(MPA)
Acetate in water pH 8.5



Mobile Phase B
5:95 MPA:ACN



(MPB)









Injection Volume
5 μL



















LC Gradient












Time
Flow
Gradient



Step
(s)
(mL/min)
Setting
% MPB














1
50
0.400
Step
5


2
30
0.400
Ramp
95


3
70
0.400
Step
5










Data is collected starting at 0.08 min and is collected over a data window length of 0.70 min.


The following MS parameters are used for analysis of the extracted samples using an API-5500 Mass Spectrometer:















Interface:
Turbo Ion Spray Ionization, positive-ion mode


Scan Mode:
Multiple Reaction Monitoring (MRM)













Parent/Product:
Dwell Time (ms):





Scan Parameters:
506.0/159.0
50



521.0/159.0
25



265.0/166.8
50



268.0/169.8
25








Source Temperature:
400° C.









The collected MS data is analyzed and sample concentrations are quantified using peak area ratios with a linear 1/x2 regression type.

Claims
  • 1. A compound 1-(5-((4-(difluoromethoxy)phenyl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2,2-dimethylpropan-1-one, or a pharmaceutically acceptable salt thereof.
  • 2. A compound 1-(5-((4-(difluoromethoxy)phenyl)sulfonyl)-3,4,5,6-tetrahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)-3-hydroxy-2,2-dimethylpropan-1-one.
  • 3. A pharmaceutical composition comprising the compound of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 4. A pharmaceutical composition comprising the compound of claim 2 and a pharmaceutically acceptable carrier.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 17/008,787, filed on Sep. 1, 2020, which is a continuation of U.S. patent application Ser. No. 16/576,720, filed on Sep. 19, 2019, which claims the benefit of U.S. Provisional Application No. 62/733,558, filed on Sep. 19, 2018, U.S. Provisional Application No. 62/733,562, filed on Sep. 19, 2018, U.S. Provisional Application No. 62/782,933, filed on Dec. 20, 2018, U.S. Provisional Application No. 62/789,641, filed on Jan. 8, 2019, and U.S. Provisional Application No. 62/811,904, filed on Feb. 28, 2019, each of which is incorporated by reference in its entirety.

US Referenced Citations (134)
Number Name Date Kind
4602093 Baldwin et al. Jul 1986 A
4918073 Ruger et al. Apr 1990 A
5030631 Bauer Jul 1991 A
5037467 Cho et al. Aug 1991 A
5059605 Clough et al. Oct 1991 A
5089621 Kim et al. Feb 1992 A
5091384 Kim et al. Feb 1992 A
5180719 White et al. Jan 1993 A
5250544 Lavielle et al. Oct 1993 A
5336772 Saiki et al. Aug 1994 A
5480899 Yano et al. Jan 1996 A
5672601 Cignarella Sep 1997 A
5714625 Hada et al. Feb 1998 A
5747502 Hanaoka et al. May 1998 A
5962703 Moszner et al. Oct 1999 A
6214879 Abraham et al. Apr 2001 B1
6534501 Abraham et al. Mar 2003 B2
6710052 Pease et al. Mar 2004 B2
6878715 Klein et al. Apr 2005 B1
7138401 Kasibhatla et al. Nov 2006 B2
7160885 Currie et al. Jan 2007 B2
7875603 Rathinavelu et al. Jan 2011 B2
8501953 Salituro et al. Aug 2013 B2
8552050 Cantley et al. Oct 2013 B2
8692001 Becker et al. Apr 2014 B2
8742119 Salituro et al. Jun 2014 B2
8785450 Salituro et al. Jul 2014 B2
8841305 Thomas et al. Sep 2014 B2
8877791 Cantley et al. Nov 2014 B2
8889667 Salituro et al. Nov 2014 B2
8952171 Xu et al. Feb 2015 B2
9012450 Metcalf et al. Apr 2015 B2
9018210 Metcalf et al. Apr 2015 B2
9108921 Cianchetta et al. Aug 2015 B2
9181231 Su Nov 2015 B2
9221792 Salituro et al. Dec 2015 B2
9248199 Metcalf Feb 2016 B2
9394257 Ho et al. Jul 2016 B2
9422279 Metcalf et al. Aug 2016 B2
9458132 Cianchetta et al. Oct 2016 B2
9458139 Xu et al. Oct 2016 B2
9604999 Harris et al. Mar 2017 B2
9708267 Boxer et al. Jul 2017 B2
9744145 Liu et al. Aug 2017 B1
9776960 Xu et al. Oct 2017 B2
9802900 Li et al. Oct 2017 B2
9957250 Metcalf et al. May 2018 B2
9981939 Metcalf et al. May 2018 B2
10004725 Dufu et al. Jun 2018 B2
10017491 Metcalf et al. Jul 2018 B2
10034879 Metcalf et al. Jul 2018 B2
10077249 Li et al. Sep 2018 B2
10100040 Li et al. Oct 2018 B2
10100043 Metcalf et al. Oct 2018 B2
10208052 Zheng et al. Feb 2019 B1
10266551 Li et al. Apr 2019 B2
10315991 Xu et al. Jun 2019 B2
10377741 Metcalf et al. Aug 2019 B2
10435393 Xu et al. Oct 2019 B2
10450269 Xu et al. Oct 2019 B1
10472371 Zheng et al. Nov 2019 B2
10493035 Dalziel et al. Dec 2019 B2
10577345 Li et al. Mar 2020 B2
10675274 Ericsson et al. Jun 2020 B2
10683285 Li Jun 2020 B2
10695330 Li et al. Jun 2020 B2
10836771 Zheng et al. Nov 2020 B2
11014927 Ericsson et al. May 2021 B2
11071725 Ericsson et al. Jul 2021 B2
11396513 Zheng et al. Jul 2022 B2
11649242 Ericsson et al. May 2023 B2
11844787 Ericsson et al. Dec 2023 B2
20040077648 Timmer et al. Apr 2004 A1
20040102458 Chiosis et al. May 2004 A1
20050002861 Krause et al. Jan 2005 A1
20050049263 Kasibhatla et al. Mar 2005 A1
20050059663 Martin et al. Mar 2005 A1
20050181305 Shibuya Aug 2005 A1
20050256103 Suzuki et al. Nov 2005 A1
20060074121 Chen et al. Apr 2006 A1
20060211737 Huang et al. Sep 2006 A1
20070015752 Hangauer, Jr. Jan 2007 A1
20070270433 Brinkman et al. Nov 2007 A1
20080058315 Cai et al. Mar 2008 A1
20080184495 Brun et al. Aug 2008 A1
20080253965 Chiosis et al. Oct 2008 A1
20080269234 Gandhi et al. Oct 2008 A1
20090042966 Coleman et al. Feb 2009 A1
20090163545 Goldfarb Jun 2009 A1
20090291921 Jabri et al. Nov 2009 A1
20100029575 Junien et al. Feb 2010 A1
20100120863 Biftu et al. May 2010 A1
20100144594 Zoller et al. Jun 2010 A1
20100144722 Alexander et al. Jun 2010 A1
20100152157 Puech et al. Jun 2010 A1
20100179154 Almario Garcia et al. Jul 2010 A1
20100216774 Bender et al. Aug 2010 A1
20100324030 Dale et al. Dec 2010 A1
20110059089 Swagemakers et al. Mar 2011 A1
20110085969 Rollo et al. Apr 2011 A1
20110104054 Chiosis et al. May 2011 A1
20120028986 Zoller Feb 2012 A1
20120134979 Xia et al. May 2012 A1
20120252818 Chiosis et al. Oct 2012 A1
20130109684 Blagg et al. May 2013 A1
20130116430 Fujiwara et al. May 2013 A1
20130155489 Kato et al. Jun 2013 A1
20130190315 Metcalf et al. Jul 2013 A1
20130190316 Metcalf et al. Jul 2013 A1
20140228360 Duncan et al. Aug 2014 A1
20140242602 Chiosis et al. Aug 2014 A1
20150246025 Desai et al. Sep 2015 A1
20160106728 Shen et al. Apr 2016 A1
20160200681 Yu et al. Jul 2016 A1
20170121338 Zhang et al. May 2017 A1
20170216434 Hines et al. Aug 2017 A1
20170217964 Li Aug 2017 A1
20180215765 Di Giorgio et al. Aug 2018 A1
20180282369 Desai et al. Oct 2018 A1
20190218221 Zheng et al. Jul 2019 A1
20200031839 Zheng et al. Jan 2020 A1
20200069643 Ericsson Mar 2020 A1
20200085798 Ericsson Mar 2020 A1
20200087309 Lancia, Jr. Mar 2020 A1
20200129485 Ericsson et al. Apr 2020 A1
20200253939 Ericsson et al. Aug 2020 A1
20210017184 Zheng et al. Jan 2021 A1
20210246143 Ericsson et al. Aug 2021 A1
20220031671 Ericsson et al. Feb 2022 A1
20220304987 Ericsson et al. Sep 2022 A1
20220378755 Luke et al. Dec 2022 A1
20220378756 Ericsson et al. Dec 2022 A1
20230055923 Zheng Feb 2023 A1
20230065368 Follows Mar 2023 A1
Foreign Referenced Citations (304)
Number Date Country
101812063 Aug 2010 CN
102206217 Oct 2011 CN
102952139 Mar 2013 CN
103570722 Feb 2014 CN
104736534 Jun 2015 CN
105037367 Nov 2015 CN
105085528 Nov 2015 CN
105153119 Dec 2015 CN
105254628 Jan 2016 CN
105294694 Feb 2016 CN
105348286 Feb 2016 CN
106928222 Jul 2017 CN
109912610 Jun 2019 CN
102008010661 Sep 2009 DE
0007529 Feb 1980 EP
0036711 Sep 1981 EP
0264883 Apr 1988 EP
0273534 Jul 1988 EP
0338372 Oct 1988 EP
0363212 Apr 1990 EP
0378255 Jul 1990 EP
0424850 May 1991 EP
0424851 May 1991 EP
0424852 May 1991 EP
0486022 May 1992 EP
0520277 Dec 1992 EP
0590415 Apr 1994 EP
0737670 Oct 1996 EP
1096310 May 2001 EP
1099692 May 2001 EP
1249233 Oct 2002 EP
1952800 Aug 2008 EP
3141542 Mar 2017 EP
2797416 Aug 2017 EP
3483164 May 2019 EP
1809MUM2013 May 2013 IN
2013MU01809 Mar 2015 IN
S 61 200544 Sep 1986 JP
3 13040 Feb 1991 JP
3 275666 Dec 1991 JP
04 054181 Feb 1992 JP
05125050 May 1993 JP
05 196976 Aug 1993 JP
7 164400 Jun 1995 JP
1 110376 Jan 1999 JP
2001261653 Sep 2001 JP
2003514673 Apr 2003 JP
2004175674 Jun 2004 JP
2007246885 Sep 2007 JP
2007328090 Dec 2007 JP
2008031064 Feb 2008 JP
2008063256 Mar 2008 JP
2009149707 Jul 2009 JP
2009212473 Sep 2009 JP
2010192782 Sep 2010 JP
2011246649 Dec 2011 JP
2012188474 Oct 2012 JP
2012188475 Oct 2012 JP
2013171968 Sep 2013 JP
20110096442 Aug 2011 KR
11379 Jul 2018 LB
2517693 Apr 2011 RU
2472794 Nov 2012 RU
WO 1993011106 Jun 1993 WO
WO 1993022298 Nov 1993 WO
WO 1995019353 Jul 1995 WO
WO 1998038239 Sep 1998 WO
WO 1998050364 Nov 1998 WO
WO 1999001442 Jan 1999 WO
WO 1999002493 Jan 1999 WO
WO 1999047489 Sep 1999 WO
WO 1999047516 Sep 1999 WO
WO 1999048461 Sep 1999 WO
WO 1999048490 Sep 1999 WO
WO 1999065895 Dec 1999 WO
WO 1999065901 Dec 1999 WO
WO 2000004023 Jan 2000 WO
WO 2000021951 Apr 2000 WO
WO 2000053591 Sep 2000 WO
WO 2001010842 Feb 2001 WO
WO 2001032764 May 2001 WO
WO 2001043744 Jun 2001 WO
WO 2001053288 Jul 2001 WO
WO 2001057037 Aug 2001 WO
WO 2001085728 Nov 2001 WO
WO 2002030358 Apr 2002 WO
WO 2002034754 May 2002 WO
WO 2002060902 Aug 2002 WO
WO 2002076989 Oct 2002 WO
WO 2002095063 Nov 2002 WO
WO 2003015769 Feb 2003 WO
WO 2003037860 May 2003 WO
WO 2003063794 Aug 2003 WO
WO 2003067332 Aug 2003 WO
WO 2003084948 Oct 2003 WO
WO 2004002490 Jan 2004 WO
WO 2004007770 Jan 2004 WO
WO 2004009600 Jan 2004 WO
WO 2004013144 Feb 2004 WO
WO 2004014374 Feb 2004 WO
WO 2004014382 Feb 2004 WO
WO 2004024676 Mar 2004 WO
WO 2004080457 Sep 2004 WO
WO 2004089470 Oct 2004 WO
WO 2004089947 Oct 2004 WO
WO 2004104000 Dec 2004 WO
WO 2005000098 Jan 2005 WO
WO 2005002577 Jan 2005 WO
WO 2005009965 Feb 2005 WO
WO 2005011653 Feb 2005 WO
WO 2005011656 Feb 2005 WO
WO 2005016915 Feb 2005 WO
WO 2005023761 Mar 2005 WO
WO 2005049570 Jun 2005 WO
WO 2005058869 Jun 2005 WO
WO 2005058870 Jun 2005 WO
WO 2005058871 Jun 2005 WO
WO 2005058873 Jun 2005 WO
WO 2005058874 Jun 2005 WO
WO 2005084667 Sep 2005 WO
WO 2005094251 Oct 2005 WO
WO 2005094834 Oct 2005 WO
WO 2005103015 Nov 2005 WO
WO 2006002100 Jan 2006 WO
WO 2006009886 Jan 2006 WO
WO 2006018279 Feb 2006 WO
WO 2006018280 Feb 2006 WO
WO 2006021448 Mar 2006 WO
WO 2006023608 Mar 2006 WO
WO 2006034315 Mar 2006 WO
WO 2006038172 Apr 2006 WO
WO 2006060122 Jun 2006 WO
WO 2006084030 Aug 2006 WO
WO 2006086445 Aug 2006 WO
WO 2006099884 Sep 2006 WO
WO 2006101521 Sep 2006 WO
WO 2006110390 Oct 2006 WO
WO 2006123121 Nov 2006 WO
WO 2006130469 Dec 2006 WO
WO 2006137485 Dec 2006 WO
WO 2007006926 Jan 2007 WO
WO 2007007069 Jan 2007 WO
WO 2007019344 Feb 2007 WO
WO 2007027734 Mar 2007 WO
WO 2007042325 Apr 2007 WO
WO 2007083119 Jul 2007 WO
WO 2007087231 Aug 2007 WO
WO 2007088123 Aug 2007 WO
WO 2007097931 Aug 2007 WO
WO 2007098418 Aug 2007 WO
WO 2007126745 Nov 2007 WO
WO 2007136603 Nov 2007 WO
WO 2007138351 Dec 2007 WO
WO 2008005937 Jan 2008 WO
WO 2008019139 Feb 2008 WO
WO 2008032905 Mar 2008 WO
WO 2008057608 May 2008 WO
WO 2008083027 Jul 2008 WO
WO 2008094203 Aug 2008 WO
WO 2008115719 Sep 2008 WO
WO 2008120003 Oct 2008 WO
WO 2008135141 Nov 2008 WO
WO 2008139585 Nov 2008 WO
WO 2009001126 Dec 2008 WO
WO 2009004356 Jan 2009 WO
WO 2009025781 Feb 2009 WO
WO 2009025784 Feb 2009 WO
WO 2009063244 May 2009 WO
WO 2009077527 Jun 2009 WO
WO 2009093032 Jul 2009 WO
WO 2009112677 Sep 2009 WO
WO 2009121623 Oct 2009 WO
WO 2009136889 Nov 2009 WO
WO 2009153554 Dec 2009 WO
WO 2010002802 Jan 2010 WO
WO 2010021717 Feb 2010 WO
WO 2010028761 Mar 2010 WO
WO 2010042867 Apr 2010 WO
WO 2010058318 May 2010 WO
WO 2010092181 Aug 2010 WO
WO 2010105243 Sep 2010 WO
WO 2010108268 Sep 2010 WO
WO 2010115688 Oct 2010 WO
WO 2010118063 Oct 2010 WO
WO 2010129596 Nov 2010 WO
WO 2010132599 Nov 2010 WO
WO 2010135524 Nov 2010 WO
WO 2010151797 Dec 2010 WO
WO 2011002816 Jan 2011 WO
WO 2011002817 Jan 2011 WO
WO 2011025690 Mar 2011 WO
WO 2011037793 Mar 2011 WO
WO 2011050210 Apr 2011 WO
WO 2011050211 Apr 2011 WO
WO 2011060321 May 2011 WO
WO 2011063055 May 2011 WO
WO 2011103256 Aug 2011 WO
WO 2011116282 Sep 2011 WO
WO 2011137089 Nov 2011 WO
WO 2011146358 Nov 2011 WO
WO 2012002577 Jan 2012 WO
WO 2012007861 Jan 2012 WO
WO 2012007868 Jan 2012 WO
WO 2012007877 Jan 2012 WO
WO 2012019426 Feb 2012 WO
WO 2012019427 Feb 2012 WO
WO 2012056319 May 2012 WO
WO 2012068096 May 2012 WO
WO 2012071519 May 2012 WO
WO 2012071684 Jun 2012 WO
WO 2012080729 Jun 2012 WO
WO 2012083246 Jun 2012 WO
WO 2012088314 Jun 2012 WO
WO 2012092426 Jul 2012 WO
WO 2012092442 Jul 2012 WO
WO 2012092485 Jul 2012 WO
WO 2012151440 Nov 2012 WO
WO 2012151448 Nov 2012 WO
WO 2012151450 Nov 2012 WO
WO 2012151451 Nov 2012 WO
WO 2012151452 Nov 2012 WO
WO 2012160392 Nov 2012 WO
WO 2012160447 Nov 2012 WO
WO 2012174126 Dec 2012 WO
WO 2013003249 Jan 2013 WO
WO 2013003250 Jan 2013 WO
WO 2013021054 Feb 2013 WO
WO 2013038390 Mar 2013 WO
WO 2013056153 Apr 2013 WO
WO 2013102142 Jul 2013 WO
WO 2013102826 Jul 2013 WO
WO 2013118805 Aug 2013 WO
WO 2013126856 Aug 2013 WO
WO 2013127266 Sep 2013 WO
WO 2013155223 Oct 2013 WO
WO 2013177224 Nov 2013 WO
WO 2013184794 Dec 2013 WO
WO 2014008458 Jan 2014 WO
WO 2014014050 Jan 2014 WO
WO 2014018355 Jan 2014 WO
WO 2014023814 Feb 2014 WO
WO 2014044356 Mar 2014 WO
WO 2014048865 Apr 2014 WO
WO 2014061031 Apr 2014 WO
WO 2014062838 Apr 2014 WO
WO 2014074848 May 2014 WO
WO 2014102817 Jul 2014 WO
WO 2014118634 Aug 2014 WO
WO 2014130890 Aug 2014 WO
WO 2014139144 Sep 2014 WO
WO 2014139325 Sep 2014 WO
WO 2014139978 Sep 2014 WO
WO 2014144715 Sep 2014 WO
WO 2014150276 Sep 2014 WO
WO 2014152588 Sep 2014 WO
WO 2014172638 Oct 2014 WO
WO 2015030514 Mar 2015 WO
WO 2015036078 Mar 2015 WO
WO 2015042397 Mar 2015 WO
WO 2015048336 Apr 2015 WO
WO 2015051230 Apr 2015 WO
WO 2015054555 Apr 2015 WO
WO 2015078374 Jun 2015 WO
WO 2015093948 Jun 2015 WO
WO 2015116061 Aug 2015 WO
WO 2015130915 Sep 2015 WO
WO 2015144605 Oct 2015 WO
WO 2015172732 Nov 2015 WO
WO 2015183173 Dec 2015 WO
WO 2015192701 Dec 2015 WO
WO 2016005576 Jan 2016 WO
WO 2016005577 Jan 2016 WO
WO 2016014324 Jan 2016 WO
WO 2016014522 Jan 2016 WO
WO 2016021815 Feb 2016 WO
WO 2016044604 Mar 2016 WO
WO 2016044629 Mar 2016 WO
WO 2016044650 Mar 2016 WO
WO 2016046837 Mar 2016 WO
WO 2016047592 Mar 2016 WO
WO 2016168647 Oct 2016 WO
WO 2016181408 Nov 2016 WO
WO 2016196816 Dec 2016 WO
WO 2016201227 Dec 2016 WO
WO 2017006270 Jan 2017 WO
WO 2017050791 Mar 2017 WO
WO 2017050792 Mar 2017 WO
WO 2017191274 Nov 2017 WO
WO 2017214002 Dec 2017 WO
WO 2018049263 Mar 2018 WO
WO 2018109277 Jun 2018 WO
WO 2018175474 Sep 2018 WO
WO 2019035863 Feb 2019 WO
WO 2019035864 Feb 2019 WO
WO 2019035865 Feb 2019 WO
WO 2019099651 May 2019 WO
WO 2019104134 May 2019 WO
WO 2019113359 Jun 2019 WO
WO 2020061252 Mar 2020 WO
WO 2020061255 Mar 2020 WO
WO 2020061261 Mar 2020 WO
WO 2020061378 Mar 2020 WO
2020139916 Jul 2020 WO
WO 2020191022 Sep 2020 WO
Non-Patent Literature Citations (375)
Entry
US 9,328,507 B2, 05/2016, Salituro et al. (withdrawn)
Schroeder; Journal of Pharmacology and Experimental Therapeutics Mar. 1, 2022, 380 (3) 210-219; DOI: https://doi.org/10.1124/jpet.121.000743 (Year: 2022).
National Center for Biotechnology Information. PubChem Substance Record for SID 377251214, SCHEMBL20511283, Source: SureChEMBL. Available Dec. 15, 2018. (Year: 2018).
SureChEMBL; “Open Patent Data”, Available Databases, 2 pages. Downloaded Oct. 31, 2023 from https://www.surechembl.org/knowledgebase/75969. (Year: 2023).
Clinical Trial Study—NCT04000165 “A Dose-Finding Study of AG-348 in Sickle Cell Disease”, ClinicalTrials.gov, Jun. 27, 2019, 9 pages.
Kalfa, T.A. et al., “Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Studies of the Safety, Tolerability, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, an Allosteric Activator of Pyruvate Kinase-R, in Healthy and Sickle Cell Disease Subjects”, Blood, American Society of Hematology, Nov. 13, 2019, pp. 3, vol. 134.
Supplemental European Search Report for EP Application 20 86 4351, dated Aug. 2, 2023, 10 pages.
Abbady M.A., et al., Synthesis and biological activity of some new 4-(2-pyrazolin-3-yl)-, 4-(2-isoxazolin-e-yl)- and 4-(1,2,5,6-tetrahydro-2-thioxopyrimidin-4-yl)phenyl aminophenyl sulfides and sulfones., Egyptian Journal of Pharmaceutical Sciences, vol. 27, No. 1-4, (1986), Abstract Only.
Abraham DJ, Mehanna AS, Wireko FC, et al. “Vanillin, a potential agent for the treatment of sickle cell anemia.” Blood. 1991;77(6):1334-41.
Adakveo [package insert]. East Hanover, New Jersey, Novartis Pharmaceuticals Corporation (Nov. 2019), 10 pgs.
Agios First Quarter 2020 Financial Results (Apr. 30, 2020), pp. 1-22.
Agrawal RK, Patel RK, Shah V, Nainiwal L, Trivedi B. “Hydroxyurea in sickle cell disease: drug review.” Indian J Hematol Blood Transfus. Jun. 2014, 30(2):91-96.
Agrawal, R. K. et al., “Hydroxyurea in Sickle Cell Disease: Drug Review”, Indian J. Hematol Blood Transfus, 30(2), pp. 91-96, (Apr.-Jun. 2014).
Aiuti, A. et al, Progress and prospects: gene therapy clinical trials (part 2), Gene Ther, 14(22): 1555-1563 (2007).
Al-Hakim, A.K. et al., 14-3-3 cooperates with LKB1 to regulate the activity and localization of QSK and SIK, Journal of Cell Science 118 (23), pp. 5661-5673 (Aug. 2005).
Al-Hakim, A.K. et al., “Control of AMPK-related kinases by USP9X and atypical Lys29/Lys33-linked polyubiquitin chains”, Biochemical Journal, 411 (2), pp. 249-260, (Feb. 2008).
Alves-Filho, J.C. & Palsson-Mcdermott, E.M., Pyruvate Kinase M2: A Potential Target for Regulating Inflammation, Frontiers in Immunology, 7(145): Article 145 (2016).
Ambrus, J. et al., Studies on the vasoocclusive crisis of sickle cell disease. III. In vitro and in vivo effect of the pyrimido-pyrimidine derivative, RA-233: studies on its mechanism of action, J Med, 18(3-4):165-198 (1987).
Amer, J. et al., Red blood cells, platelets and polymorphonuclear neutrophils of patients with sickle cell disease exhibit oxidative stress that can be ameliorated by antioxidants, British Journal of Haematology, 132(1):108-113 (2006).
Andresen, C.A. et al., “Protein Interaction Screening for the Ankyrin Repeats and Suppressor of Cytokine Signaling (SOCS) Box (ASB) Family Identify Asb11 as a Novel Endoplasmic Reticulum Resident Ubiquitin Ligase”, The Journal of Biological Chemistry, vol. 289, No. 4, pp. 2043-2054, (Jan. 24, 2014).
Ataga KI, Kutlar A, Kanter J, Liles D, Cancado R, Friedrisch J, Guthrie TH, Knight-Madden J, Alvarez OA, Gordeuk VR, Gualandro S, Colella MP, Smith WR, Rollins SA, Stocker JW, Rother RP. “Crizanlizumab for the prevention of pain crises in sickle cell disease.” N Engl J Med. Feb. 2, 2017, 376(5):429-439.
Atkinson, Peter J., et al., 3,4-Dihydro-2H-benzoxazinones are 5-HT1A receptor antagonists with potent 5-HT reuptake inhibitory activity, BioOrganic & Medicinal Chemistry Letters, 15(3), pp. 737-741 (2005).
Austin, Nigel E., et al., “Novel 2,3,4,5-tetrahydro-1H-3-benzazepines with high affinity and selectivity for the dopamine D3 receptor”, BioOrganic & Medicinal Chemistry Letters, 10(22), pp. 2553-2555, (2000).
Bailey, S.D. et al., “Variation at the NFATC2 Locus Increases the Risk of Thiazolidinedione-Induced Edema in the Diabetes Reduction Assessment with Ramipril and rosiglitazone Medication (DREAM) Study”, Diabetes Care, vol. 33, No. 10, pp. 2250-2254, (Oct. 2010).
Bakshi N, Sinha CB, Ross D, Khemani K, Loewenstein G, Krishnamurti L. “Proponent or collaborative: Physician perspectives and approaches to disease modifying therapies in sickle cell disease.” PLoS One. Jul. 20, 2017, 12(7):e0178413.
Balakin, Konstantin V. et al., Comprehensive Computational Assessment of ADME Properties using Mapping Techniques, Current Drug Discovery Technologies, 2(2), pp. 99-113 (2005).
Banerjee, S. et al., “Interplay between Polo kinase, LKB1-activated NUAK1 kinase, PP1β phosphatase complex and the SCFβTrCP E3 ubiquitin ligase”, Biochem. J. 461, pp. 233-245, (2014).
Banerjee, T. and Kuypers F.A., Reactive oxygen species and phosphatidylserine externalization in murine sickle red cells, British Journal of Haematology, 124:391-402 (2004).
Barbier AJ, Bodie S, Connor G, et al. “Safety, tolerability, pharmacokinetics and pharmacodynamics of multiple doses of AG-519, an allosteric activator of pyruvate kinase-R, in healthy subjects.” Blood. 2016, 128:1264.
Barua, A.K., et al., Chemistry and Industry Communications to the Editor 1376 24 (Oct. 1970).
Bennett, Eric J., et al., “Dynamics of Cullin-RING Ubiquitin Ligase Network Revealed by Systematic Quantitative Proteomics”, Cell 143, pp. 951-965, (Dec. 10, 2010).
Betz T, Lenz M, Joanny JF, Sykes C. “ATP-dependent mechanics of red blood cells.” Proc Natl Acad Sci USA. 2009;106(36):15320-5.
Beutler, E. and Gelbart, T., “Estimating the prevalence of pyruvate kinase deficiency from the gene frequency in the general white population”, Blood, 95(11): 3585-3588 (2000).
Bianchi, P. and Zanella, A., “Hematologically important mutations: red cell pyruvate kinase”, (Third update), Blood Cells Mol Dis., 26(1): 47-53 (2000).
Biftu, T. et al., “Omarigliptin (MK-3102): A Novel Long-Acting DPP-4 Inhibitor for Once-Weekly Treatment of Type 2 Diabetes”, Journal of Medicinal Chemistry, 57, pp. 3205-3212, (2014).
Bouwmeester, T. et al., “A physical and functional map of the human TNF-α/NF-κB signal transduction pathway”, Nature Cell Biology, vol. 6, No. 2, pp. 97-105, (Feb. 2004).
Boxer, M.B. et al., “Evaluation of Substituted N,N1-Diarylsulfonamides as Activators of the Tumor Cell Specific M2 Isoform of Pyruvate Kinase”, J. Med. Chem., 53: pp. 1048-1055 (2010).
Brajenovic, M. et al., “Comprehensive Proteomic Analysis of Human Par Protein Complexes Reveals an Interconnected Protein Network”, The Journal of Biological Chemistry, vol. 275, No. 13, pp. 12804-12811 (Mar. 2004).
Brehme, M. et al., “Charting the molecular network of the drug target Bcr-Abl”, PNAS, vol. 106, No. 18, pp. 7414-7419, (May 2009).
Bridges, C.R., et al., “USP9X deubiquitylating enzyme maintains RAPTOR protein levels, mTORC1 signalling and proliferation in neural progenitors”, Scientific Reports 7:391, pp. 1-15, (Mar. 2017).
Brown, R. Clark, et al., “FT-4202, an Allosteric Activator of Pyruvate Kinase-R, Demonstrates Proof of Mechanism and Proof of Concept after a Single Dose and after Multiple Daily Doses in a Phase 1 Study of Patients with Sickle Cell Disease,” Blood (2020) 136 (Supplement 1):19-20, Nov. 4, 2020.
Brown, R. Clark, et al., “FT-4202, an Allosteric Activator of Pyruvate Kinase-R, Demonstrates Proof of Mechanism and Proof of Concept after a Single Dose and after Multiple Daily Doses in a Phase 1 Study of Patients with Sickle Cell Disease,” ASH 2020, Dec. 7, 2020.
Budzikiewicz, Herbert et al., “Vincetene, a benzopyrroloisoquinoline alkaloid, from Cynanchum vincetoxicum (L.) Pers. (Asclepiadaceae)”, Liebigs Annalen Der Chemie, (8), pp. 1212-1231 (1979).
Buontempo P, Jubin RG, Buontempo C, Real R, Kazo F, O'Brien S, Adeel F, Abuchowski A. “Pegylated carboxyhemoglobin bovine (SANGUINATE) restores RBCs roundness and reduces pain during a sickle cell vaso-occlusive crisis.” Blood. 2017, 130:969.
Cabrales, P. et al., “A look inside the mechanistic black box: Are red blood cells the critical effectors of RRx-001 cytotoxicity?”, Med Oncol., 33(7):63 (2016).
CAS Registry No. 1208929-16-1, Tert-Butyl 1H,2H,3H,4H,5H,6H-Pyrrolo[3,4-C]Pyrrole-2-Carboxylate Hydrochloride (Mar. 11, 2010).
Castilhos, L. et al., “Altered E-NTPDase/E-ADA activities and CD39 expression in platelets of sickle cell anemia patients”, Biomed Pharmacother., 79:241-246 (2016).
Castilhos, L. et al., “Increased oxidative stress alters nucleosides metabolite levels in sickle cell anemia”, Redox Rep., 22(6):451-459 (2017).
Castilhos, L. et al., “Sickle cell anemia induces changes in peripheral lymphocytes E-NTPDase/E-ADA activities and cytokines secretion in patients under treatment”, Biomedicine & Pharmacotherapy 73 (2015), pp. 102-108.
Castro, O., Viability and function of stored sickle erythrocytes, Transfusion, 20(6):695-703 (1980).
Cazzola, M., Pyruvate kinase deficiency, Haematologica, 90(1): 1-2 (2005).
Charache, S. et al., Effect of 2,3-Diphosphateglycerate on oxygen affinity of blood in sickle, Cell Anemia, Journal ofClinical Investigation, 49(4):806-812 (1970).
Chaudhary, Neelam & Maddika, Subbareddy, “WWP2-WWP1 Ubiquitin Ligase Complex Coordinated by PPM1G Maintains the Balance Between Cellular p73 and AΔNp73 Levels”, Mol. Cell. Biol. (Oct. 2014).
Chen, Yue et al.—Preclinical Pharmacokinetic/Pharmacodynamic Relationships for AG-348, An Investigational Small-Molecule Activator of Pyruvate Kinase, European Hematology Association, Jun. 13, 2015.
Cheung, Yiu-Yin et al., Solution-Phase Parallel Synthesis and SAR of Homopiperazinyl Analogs as Positive Allosteric Modulators of MGlu4, ACS Comb Sci. 13(2), pp.159-165, (Mar. 2011).
Chiosis et al., Development of a Purine-Scaffold Novel Class of Hsp90 Binders that Inhibit the Proliferation of Cancer Cells and Induce the Degradation of Her2 Tyrosine Kinase, BioOrganic & Medicinal Chemistry, vol. 10, Iss 11, (Nov. 2002), pp. 3555-3564.
Chiou WL, Barve A. “Linear correlation of the fraction of oral dose absorbed of 64 drugs between humans and rats.” Pharm Res. Nov. 1998, 15(11):1792-5.
Chonat, S. et al.,—Improvement in Red Blood Cell Physiology in Children With Sickle Cell Anemia Receiving Voxelotor—Childrens Healthcare of Atlanta (Dec. 2019).
Choudhury, N.R., et al., “RNA-binding activity of TRIM25 is mediated by its PRY/SPRY domain and is required for ubiquitination”, BMC Biology 15:105, pp. 1-20, (2017).
Christensen, R.D. et al., Siblings with Severe Pyruvate Kinase Deficiency and a Complex Genotype, American Journal of Medical Genetics, Part A, (2016), pp. 2449-2452.
Chubukov V, Johnson K, Kosinski PA, et al. “Characterization of metabolic response to AG-348, an allosteric activator of red cell pyruvate kinase, in healthy volunteers and pyruvate kinase deficiency patients.” Poster presented at: 58th American Society of Hematology Annual Meeting and Exposition; Dec. 4, 2016; San Diego, California. http://investor.agios.com/staticfiles/e1e9fd70-c84b-4472-bff3-bef0ecf05482 Accessed Jul. 28, 2017.
Chung, J.Y.L. et al., “Evolution of a Manufacturing Route to Omarigliptin, A Long-Acting DPP-4 Inhibitor for the Treatment of Type 2 Diabetes”, Organic Process Research & Development, 19, pp. 1760-1768, (2015).
Clinical Trial Study—NCT02604433—U.S. National Library of Medicine, An Efficacy and Safety Study of Luspatercept (ACE-536) Versus Placebo in Adults Who Require Regular Red Blood Cell Transfusions Due to Beta (β) Thalassemia (BELIEVE), Submitted Date: Nov. 13, 2015, 24 pgs.
ClinicalTrlals.gov, NCT03815695, (v1)—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients” (Jan. 22, 2019).
Clinical Trials Study, NCT03815695, (v2)—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” (Mar. 13, 2019) pp. 1-5.
ClinicalTrlals.gov, NCT03815695, (v3)—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients” (Sep. 16, 2019).
Clinical Trial Study—NCT03815695, (v4)—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Sep. 19, 2019 (v4), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Sep. 23, 2019 (v5), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Oct. 9, 2019 (v6), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Oct. 10, 2019 (v7), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Nov. 27, 2019 (v8), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Jan. 15, 2020 (v9 ), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Jan. 16, 2020 (v10), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Date: Feb. 21, 2020 (v11), 5 pgs.
Clinical Trial Study NCT03815695—U.S. National Library of Medicine, “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Disease Patients,” Submitted Apr. 1, 2020, (v12), 12 pgs.
ClinicalTrials.gov, NCT03815695 (v13), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Jun. 15, 2020.
ClinicalTrials.gov, NCT03815695 (v14), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Jul. 17, 2020.
ClinicalTrials.gov, NCT03815695 (v15), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Aug. 19, 2020.
ClinicalTrials.gov, NCT03815695 (v16), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Sep. 1, 2020.
ClinicalTrials.gov, NCT03815695 (v17), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Sep. 18, 2020.
ClinicalTrials.gov, NCT03815695 (v18), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamics of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Oct. 15, 2020.
ClinicalTrials.gov, NCT03815695 (v19), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamices of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Study Record Versions 19—Dec. 24, 2020.
ClinicalTrials.gov, NCT03815695 (v20), “A SAD/MAD to Assess the Safety, Pharmacokinetics and Pharmacodynamices of FT-4202 in Healthy Volunteers and Sickle Cell Patients,” Study Record Versions 20, Jan. 8, 2021.
ClinicalTrials.gov, NCT04624659 (v1), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Version 1—Nov. 5, 2020.
ClinicalTrials.gov, NCT04624659 (v2), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Version 2—Nov. 10, 2020.
ClinicalTrials.gov, NCT04624659 (v3), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Versions 3—Dec. 10, 2020.
ClinicalTrials.gov, NCT04624659 (v4), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Versions 4, Dec. 28, 2020.
ClinicalTrials.gov, NCT04624659 (v5), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Versions 5, Jan. 7, 2021.
ClinicalTrials.gov, NCT04624659 (v6), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Versions 6, Jan. 14, 2021.
ClinicalTrials.gov, NCT04624659 (v7), “A Study of FT-4202 in Adults and Adolescents With Sickle Cell Disease,” Study Record Versions 7, Feb. 8, 2021.
Cloutier, P. et al., “R2TP/Prefoldin-like component RUVBL1/RUVBL2 directly interacts with ZNHIT2 to regulate assembly of U5 small nuclear ribonucleoprotein”, Nature Communications, pp. 1-14 (May 2017).
Cole, D.C. et al., Conformationally Constrained N1-arylsulfonyltryptamine derivatives as 5-HT6 receptor antagonists, BioOrganic & Medicinal Chemistry Letters, vol. 15, No. 21, (Nov. 1, 2005), pp. 4780-4785.
Cox, J.L., et al., “The SOX2-Interactome in Brain Cancer Cell Identifies the Requirement of MSI2 and USP9X for the Growth of Brain Tumor Cell”, PLOS ONE, vol. 8, Issue 5, pp. 1-13, (May 2013).
Croasdell, G., European Hematology Association—20th Annual Congress (Jun. 11-14, 2015—Vienna, Austria) Meeting Report, Drugs of Today (2015), 51(7),I pp. 441-445.
Das, A. et al., “USP9X counteracts differential ubiquitination of NPHPS by MARCH7 and BBS11 to regulate ciliogenesis”, PLOS Genetics, pp. 1-24, (May 12, 2017).
Davis, Z.H., et al., “Global Mapping of Herpesvirus-Host Protein Complexes Reveals a Transcription Strategy for Late Genes”, Molecular Cell 57, pp. 349-360; (Jan. 22, 2015).
De Furia, F. et al., The effects of cyanate in vitro on red blood cell metabolism and function in sickle cell anemia, J Clin Invest., 51(3):566-574 (1972).
De Jong, K. and Kuypers, F., Sulphydryl modifications alter scramblase activity in murine sickle cell disease, British Journal of Haematology, 133(4):427-432 (2006).
De Rosa MC, Carelli Alinovi C, Galtieri A, Russo A, Giardina B. “Allosteric properties of hemoglobin and the plasma membrane of the erythrocyte: New insights in gas transport and metabolic modulation.” IUBMB Life. 2008, 60(2):87-93.
Diez, A. et al., Life-threatening nonspherocytic hemolytic anemia in a patient with a null mutation in the PKLR gene and no compensatory PKM gene expression, Blood, 106:1851 (2005).
Diez-Silva M, Dao M, Han J, Lim CT, Suresh S. “Shape and biomechanical characteristics of human red blood cells in health and disease.” MRS Bull. May 2010, 35(5):382-8.
Drissi, R. et al., “Quantitative Proteomics Reveals Dynamic Interactions of the Mini chromosome Maintenance Complex (MCM) in the Cellular Response to Etoposide Induced DNA Damage”, Molecular & Cellular Proteomics, pp. 2002-2013, (2015).
DROXIA [package insert]. Princeton, New Jersey, Bristol-Myers Squibb Company, (Dec. 2017), 28 pgs.
DROXIA [package insert]. Princeton, New Jersey, Bristol-Myers Squibb Company (Dec. 2019), 25 pgs.
Dupont, S. et al., “FAM/USP9x, a Deubiquitinating Enzyme Essential for TGFβ Signaling, Controls Smad4 Monoubiquitination”, Cell, 136, pp. 123-135, (Jan. 9, 2009).
Dzandu JK, Johnson RM. “Membrane protein phosphorylation in intact normal and sickle cell erythrocytes.” J Biol Chem. Jul. 10, 1980, 255(13):6382-6.
El-Sharief, A.M., et al., Some halogenated sulfonamides with biological interest, Journal of the Indian Chemical Society, vol. 61, No. 6, (1984), pp. 537-543.
Emam, H.A., et al., Heterocyclization of sulfamido chalcones to pyrazoline, cyanopyridone, nicotinonitrile and hydrobenzo [1,2-c] pyrazole derivatives, Journal of the Serbian Chemical Society, vol. 62, No. 7, (1997), Abstract only.
ENDARI [package insert]. Torrance, California: Emmaus Medical, Inc., (Jul. 2017), 8 pgs.
ENDARI [package insert]. Torrance, California, Emmaus Medical, Inc., (Nov. 2019), 10 pgs.
Ernst, A. et al., “A Strategy for Modulation of Enzymes in the Ubiquitin System”, Science, 339, pp. 1-15, (Feb. 2013).
Estepp, et al., Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Study of the Safety, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, A PKR-Activator, In Healthy and Sickle Cell Disease Subjects, Abstract, e-Poster, European Hematology Association Open Access Library, Presentation EHA25, (May 14, 2020), 2 pgs.
Estepp, et al., Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Study of the Safety, Phyarmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, a PKR Activator, in Healthy and Sickle Cell Disease Subjects, Poster, EP1531, (Jun. 12, 2020), 1 pg.
Estepp, J.H. et al., A clinically meaningful fetal hemoglobin threshold for children with sickle cell anemia during hydroxyurea therapy, Am J Hematol., 92:1333-1339 (2017).
Estepp, Jeremie H., et al., “Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Study of the Safety, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, a PKR Activator, in Healthy Volunteers and Patients with Sickle Cell Disease,” Virtual meeting [poster EP1531] presented at the 25th Congress of the European Hematology Association; Jul. 11-21, 2020.
European Hematology Association HemaSphere Abstract Book, 15th Annual Sickle Cell & Thalassaemia & 1st EHA European Sickle Cell Conference, Oct. 26-31, 2020.
Fioravanti, R., et al., Synthesis and Biological Evaluation of N-substituted-3, 5-diphenyl—2-pyrazoline derivatives as cyclooxygenase (COX-2) inhibitors, European Journal of Medicinal Chemistry, vol. 45, No. 12, (Dec. 1, 2010), pp. 6135-6138, XP027526583.
Fitch, R. W. et al., Phantasmidine: An Epibatidine Congener from the Ecuadorian Poison Frog Epipedobates anthonyi, Journal of Natural Products (2010), vol. 73, No. 3, pp. 331-337.
Fleischhacker, W., et al., “Heterocyclic fused naphthalene systems from thebaine. 1”, Liebigs Annalen Der Chemie, (5), pp. 844-851, (1983).
Fogeron, M.L. et al., “LGALS3BP regulates centriole biogenesis and centrosome hypertrophy in cancer cells”, Nature Communications, 4:1531, pp. 1-14; (2013).
Forma Therapeutics, Press Release, “Forma Therapeutics Presents Clinical Proof-of-Concept Data at the 62nd Annual ASH Meeting Supporting the Potential of its Novel Investigational PKR Activator, FT-4202, to Treat Sickle Cell Disease (SCD)” (Dec. 7, 2020).
Forma Therapeutics, Inc., Press Release—“Forma Therapeutics Announces Positive FT-4202 600 mg Multiple Ascending Dose Cohort Data Supporting the Doses Being Evaluated in Phase 2/3 Registrational Trial, Called the Hibiscus Study”, Mar. 30, 2021—2 pgs.
Frost, David A., et al., “Naturally occurring compounds related to phenalenone. V. Synthetic approaches to structures based on 8,9-dihydro-8,8,9-trimethylphenaleno [1,2-b] furan-7-one”, Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999), pp. 2159-2169.
Gaudet, P. et al., “Phylogenetic-based propagation of functional annotations within the Gene Ontology consortium”, vol. 12, No. 5, pp. 449-462; (Aug. 2011).
Giannone, R.J., et al., “The Protein Network Surrounding the Human Telomere Repeat Binding Factors TRF1, TRF2, and POT1”, PLOS One, vol. 5, Issue 8, pp. 1-10, (Aug. 2010).
Gizi, A. et al., Assessment of oxidative stress in patients with sickle cell disease: The glutathione system and the oxidant-antioxidant status, Blood Cells Mol Dis., 46(3):220-225 (2011).
Gladwin, M., Adenosine recepter crossroads in sickle cell disease, Nature Medicine, 17(1):38-40, (2011).
Glombitza, S. et al., Adenosine causes cAMP-dependent activation of chick embryo red cell carbonic anhydrase and 2,3-DPG synthesis, American Journal of Physiology, 271(4):973-81 (1996).
Gomez-Bougie, P. et al., “Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction”, Biochemical and Biophysical Research Communications 413, pp. 460-464, (2011).
Goncharov, T. et al., “OTUB1 modulates c-IAP1 stability to regulate signaling pathways”, The EMBO Journal 32, No. 8, pp. 1103-1114, (2013).
Grace RF, Rose C, Layton DM, Yaish HM, Barcellini W, Galactéros F, Morton DH, Ravindranath Y, Kuo KHM, van Beers EJ, Kwiatkowski JL, Silver BA, Merica E, Kung C, Cohen M, Yang H, Hixon J, Kosinski PA, Silver L, Dang L, Yuan Z, Barbier AJ, Glader B. “Effects of AG_348, a pyruvate kinase activator, on anemia and hemolysis in patients with pyruvate kinase deficiency: Data from the DRIVE PK study”. Blood. 2016, 128:2402.
Grace, et al., Safety and Efficacy of Mitapivat in Pyruvate Kinase Deficiency, N. Engl. J. Med. 381, 10, (Sep. 5, 2019), p. 933-944.
Grasso, D. et al., “Zymophagy, a Novel Selective Autophagy Pathway Mediated by VMPl-USP9x-p62, Prevents Pancreatic Cell Death”, The Journal of Biological Chemistry, vol. 286, No. 10, pp. 8308-8324, (Mar. 2011).
Greco, T.M. et al., “Nuclear Import of Histone Deacetylase 5 by Requisite Nuclear Localization Signal Phosphorylation”, Molecular & Cellular Proteomics 10: , pp. 1-15, (2011).
Grou, C.P., et al., “Identification of ubiquitin-specific protease 9X (USP9X) as a deubiquitinase acting on the ubiquitin-peroxin 5 (PEXS) thioester conjugate”, J. Biol. Chem., pp. 1-24; (Feb. 27, 2012).
Habata, S. et al., “BAG3-mediated Mcl-1 stabilization contributes to drug resistance via interaction with USP9X in ovarian cancer”, International Journal of Oncology 49: pp. 402-410, (2016).
Han, K.J. et al., “Ubiquitin-specific Protease 9x Deubiquitinates and Stabilizes the Spinal Muscular Atrophy Protein—Survival Motor Neuron”, J. Biol. Chem., pp. 1-22, (Oct. 2012).
Hanson, D. et al., “Identifying biological pathways that underlie primordial short stature using network analysis”, Journal of Molecular Endocrinology, pp. 333-344, (2014).
Harada, R. et al., “Structure of pristimerine, a quinonoid triterpene”, Tetrahedron Letters, pp. 603-607, (1962).
Harayama, Takashi et al., “Novel synthesis of naphthobenzazepines from N-bromobenzylnaphthylamines by regioselective C—H activation utilizing the intramolecular coordination of an amine to Pd”, Synlett, (8), pp. 1141-1144, (2003)
Hauri, S. et al., “Interaction proteome of human Hippo signaling: modular control of the co-activator YAP1”, Molecular Systems Biology, 9: 713, pp. 1-16 (Nov. 2013).
Havugimana, P. et al., “A Census of Human Soluble Protein Complexes”, Cell 150, pp. 1068-1081, (Aug. 2012).
Hebbel RP, Eaton JW, Balasingam M, Steinberg MH. “Spontaneous oxygen radical generation by sickle erythrocytes.” J Clin Invest. 1982, 70(6):1253-9.
Hein, M.Y., et al., “A Human Interactome in Three Quantitative Dimensions Organized by Stoichiometries and Abundances”, Cell 163, pp. 712-723, (Oct. 2015).
Hierso, R. et al., Effects of oxidative stress on red blood cell rheology in sickle cell patients, British Journal of Haematology, 166(4):601-606 (2014).
Homan, C.C. et al., “Mutations in USP9X are Associated with X-linked Intellectual Disability and Disrupt Neuronal Cell Migration and Growth”, The American Journal of Human Genetics 94, pp. 470-478, (Mar. 2014).
Hoppe CC, Inati AC, Brown C, et al. “Initial results from a cohort in a phase 2a study (GBT440-007) evaluating adolescents with sickle cell disease treated with multiple doses of GBT440, a HbS polymerization inhibitor.” Blood. 2017:130(Suppl 1): 689.
Husain, M.I., et al., Synthesis of some new N-[4-(acetyl/phenyl-5-arylpyrazolin-3-yl)phenyl]arylsulfonamides as oral hypoglycemic agents, Indian Drugs, vol. 24, No. 4, (1987), Abstract only.
Huttlin, E. L., et al., “The BioPlex Network: A Systematic Exploration of the Human Interactome”, Cell 162, pp. 425-440, (Jul. 2015).
Huttlin, E.L., et al., “Architecture of the human interactome defines protein communities and disease networks”, Nature, pp. 1-35, (May 2017).
HYDREA [package insert]. Princeton, NewJersey, Bristol-Myers Squibb Company (Jul. 2019), 29 pgs.
Imamura K, Tanaka T. “Multimolecular forms of pyruvate kinase from rat and other mammalian tissues. | Electrophoretic studies.” J Biochem. 1972, 71:1043-51.
Imamura K, Tanaka T. “Pyruvate kinase isozymes from rat.” Methods Enzymol. 1982, 90:150-65.
International Search Report and Written Opinion for PCT/US2019/051831, dated Dec. 6, 2019 (Dec. 6, 2020).
International Search Report and Written Opinion for PCT/US2020/051645, dated Dec. 7, 2020 (Dec. 7, 2020).
International Search Report and Written Opinion for PCT/US2020/051579, dated Dec. 10, 2020 (Dec. 10, 2020).
International Search Report and Written Opinion for PCT/US2019/052024, dated Dec. 23, 2019 (Dec. 23, 2019).
International Search Report and Written Opinion for PCT/US2018/023405, dated Jun. 5, 2018 (Jun. 5, 2018).
Iwasaki, Tameo et al., “Novel Selective PDE IV Inhibitors as Antiasthmatic Agents. Synthesis and Biological Activities of a Series of 1-Aryl-2,3-bis (hydroxymethyl) naphthalene Lignans”, Journal of Medicinal Chemistry (1996), pp. 2696-2704.
Jendralla, H. et al., Synthesis of 1,2,3,4,5,6-Hexahydropyrrolo[3,4-c]pyrrole dihydrobromide and 1,2,3,5-Tetrahydro-2-[(4-Methyl-Phenyl)Sulfonyl]Pyrrolo[3,4-c]Pyrrole, Heterocycles, 41(6): 1291-1298 (1995).
Jin, Y. et al., Effects of gamma irradiation on red cells from donors with sickle cell trait, Transfusion, 37(8):804-808 (1997).
Johansen, L.D., et al., “IKAP localizes to membrane ruffles with filamin A and regulates actin cytoskeleton organization and cell migration”, Journal of Cell Science 121, pp. 854-864, (Dec. 2007).
Jones, M.H., et al., “The Drosophila developmental gene fat facets has a human homologue in Xp11.4 which escapes X-inactivation and has related sequences on Yq11.2”, Human Molecular Genetics, vol. 5, No. 11, pp. 1695-1701, (Aug. 1996).
Jorgensen, Eugene C., et al., “Thyroxine analogs. 20. Substituted 1-and 2-naphthyl ethers of 3,5-diiodotyrosine”, Journal of Medicinal Chemistry 14(11), pp. 1023-1026, (1971).
Joshi, B., et al., Indian J. Chem., Sect. B (1983), 228(2), Abstract only. Chemical Abstract No. 99:105146.
Joshi, P., et al., “The functional interactome landscape of the human histone deacetylase family”, Molecular Systems Biology 9, 672, (2013).
Kalai, T. et al., Synthesis of Pyrroline Nitroxide Annulated Carbocycles and Heterocycles, Synthesis No. 6, pp. 831-837 (2000).
Kalfa, et al., FORMA Therapeutics, Inc., Watertown, MA, Power Pointe Presentation, Dated Nov. 6, 2019 , Phase 1 Single and Multiple Ascending Dose Study of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of FT-4202, an Allosteric activator of Pyruvate Kinase-R, in Healthy and Sickle Cell Disease Subjects, 15 pgs.
Kalfa, T. A. et al., “Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Study of the Safety, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, a PKR Activator, in Healthy and Sickle Cell Diseases Subjects”, JSCDH-D-20-0053, vol. VII, Pub. Date: Jun. 12, 2020; pp. 83-84.
Kalfa, T. et al., “Phase 1 Single (SAD) and Multiple Ascending Dose (MAD) Study of the Safety, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, a PKR Activator, in Healthy and Sickle Cell Diseases Subjects”, 14th Annual Sickle Cell Disease Research and Educational Symposium/43rd National Sickle Cell Disease Scientific Meeting (Sep. 23-25, 2020).
Kalfa, T.A. et al., “616 Phase 1 Single (SAD) and Julotiple Ascending Dose (MAD) Studies of the Safety, Tolerability, Pharmacokinetics (PK) and Pharmacodynamics (PD) of FT-4202, an Allosteric Activator of Pyruvate Kinase-R, in Healthy and Sickle Cell Disease Subjects”, (Nov. 2019).
Kaltenbach, L.S., et al., “Huntingtin Interacting Proteins are Genetic Modifiers of Neurodegeneration”, PLOS Genetics, vol. 3, Issue 5, pp. 689-708, (May 2007).
Kasturi, Tirumalai R., et al., “Reactions of tetrahalo-1,2-benzoquinones. III. Reaction of tetrachloro-1,2-benzoquinone withtetralones and naphthols: pathway to the condensates”, Journal of the Chemical Society C: Organic, (9), pp. 1257-1259, (1970).
Katzenellenbogen, R.A., et al., “NFX1-123 and Poly(A) Binding Proteins Synergistically Augment Activation of Telomerase In Human Papillomavirus Type 16 E6-Expressing Cells”, Journal of Virology, vol. 81, pp. 3786-3796, (Apr. 2007).
Khafagy, M.M., Synthesis of some pyrimidine and pyrazoline derivatives, Al-Azhar Bulletin of Science, vol. 3, No. 1, (1992), Abstract only.
Kharalkar, S.S. et al., Identification of Novel Allosteric Regulators of Human-Erythrocyte Pyruvate Kinase, Chemistry & Biodiversity, vol. 4, pp. 2603-2617 (Feb. 2007).
Kim H, Kosinski P, Kung C, Dang L, Chen Y, Yang H, Chen YS, KramerJ, Liu G. “A fit-for-purpose LC-MS/MS method for the simultaneous quantitation of ATP and 2,3-DPG in human KZEDTA whole blood.” J Chromatogr B Analyt Technol Biomed Life Sci. Sep. 1, 2017, 1061-1062:89-96.
Kim J, Lee H, Shin S. “Advances in the measurement of red blood cell deformability: A brief review.” J Cell Biotech. 2015;1263-79.
Kim, M., et al., “Role of Angiomotin-like 2 mono-ubiquitination on YAP inhibition”, EMBO reports, vol. 17, No. 1., pp. 64-78, (Nov. 23, 2015).
Kimura, K., et al., “Diversification of transcriptional modulation: Large-scale identification and characterization of putative alternative promoters of human genes”, Genome Research 16, pp. 55-65, (2006).
Kirli, K., et al., “A deep proteomics perspective on CRM1-mediated nuclear export and nucleocytoplasmic partitioning”, eLife, pp. 1-28; (2015).
Knauff, E.A.H., et al., “Genome-wide association study in premature ovarian failure patients suggests ADAMTS19 as a possible candidate gene”, Human Reproduction, vol. 24, No. 9, pp. 2372-2379, (2009).
Kodama, K. et al., Solvent-induced dual chirality switching in the optical resolution of tropic acid via diastereomeric salt formation with (1R,2S)-2-amino-1,2-diphenylethanol, Tetrahedron 70:7923-7928 (2014).
Konstantinidis, Diamantis G., et al., “Ex-Vivo FT-4202 Treatment Improves Hemoglobin Oxygen Affinity and Membrane Health in Red Blood Cells of Patients with Hemoglobin SS and Hemoglobin SC Disease Irrespective of Prior Hydroxyurea Use,” Blood (2020) 136 (Supplementl):23-24, Nov. 4, 2020.
Konstantinidis, Diamantis G., et al., “Ex-Vivo FT-4202 Treatment Improves Hemoglobin Oxygen Affinity and Membrane Health in Red Blood Cells of Patients with Hemoglobin SS and Hemoglobin SC Disease Irrespective of Prior Hydroxyurea Use,” Presented at the 62nd American Society of Hematology (ASH) Annual Meeting, Dec. 5, 2020.
Kristensen, A.R., Gsponer, J. and Foster, L.J., “A high-throughput approach for measuring temporal changes in the interactome”, Nat Methods, 9(9), pp. 1-12, (2012).
Kuehl, G. et al., In vitro interactions of 51Cr in human red blood cells and hemolysates, Vox Sang., 40(4):260-272 (1981).
Kung C, Hixon J, Kosinski PA, et al. “AG-348 enhances pyruvate kinase activity in red blood cells from patients with pyruvate kinase deficiency.” Blood. 2017;130(11):1347-1356.
Kurita, R. et al., Establishment of Immortalized Human Erythroid Progenitor Cell Lines Able to Produce Enucleated Red Blood Cells, PLOS ONE, vol. 8, Iss. 3, pp. 1-15 (Mar. 2013).
Kushwaha, D., et al., “USP9X inhibition promotes radiation-induced apoptosis in non-small cell lung cancer cells expressing mid-to-high MCL1”, Cancer Biology & Therapy 16:3, pp. 392-401, (Mar. 2015).
Kwasna, D., et al., “Discovery and Characterization of ZUFSP/ZUP1, a Distinct Deubiquitinase Class Important for Genome Stability”, Molecular Cell 70, pp. 150-164, (2018).
Le Quesne, P.W. et al., One-Step Preparation of Tetrakis(bromomethyl)ethylene from Pinacolyl Alcohol, J. Org. Chem., 40(1): 142-143 (1975).
Le, Kha et al., Population pharmacokinetics and pharmacodynamics of AG-519, a pyruvate kinase activator for the treatment of pyruvate kinase deficiency, in human healthy volunteers, Agios Pharma—1263 Poster,—58th American Society of Hematology Annual Meeting and Exposition, Dec. 3-6, 2016—San Diego, CA.
Le, Kha et al., Population pharmacokinetics and pharmacodynamics of AG-348 in healthy human volunteers guide dose selection for the treatment of pyruvate kinase deficiency, Agios Pharma—3336 Poster,—57th American Society of Hematology Annual Meeting and Exposition, Dec. 5-8, 2015—Orlando, FL.
Lehrer-Graiwer J, Howard J, Hemmaway CJ, et al. “Long-term closing in sickle cell disease subjects with GBT440, a novel HbS polymerization inhibitor.” Blood, 2016:128(22): 2488.
Lehrer-Graiwer, Josh et al., Long-Term Dosinig in Sickle Cell Disease Subjects with GBT440, a Novel HbS Polymerization Inhibitor, blood, 114, Hemoglobinopathies, Excluding Thalassemia—Clinical Poster II, Dec. 2, 2016.
Lenihan, J.A., Saha, Orthis, and Young P.W., “Proteomic analysis reveals novel ligands and substrates for LNX1 E3 ubiquitin ligase”, PLOS ONE, pp. 1-18; (Nov. 2017).
Li, X., et al., “Defining the protein-protein interaction network of the human protein tyrosine phosphatase family”, The American Society for Biochemistry and Molecular Biology, Inc., pp. 1-54, (2016).
Litinov RI, Weisel JW. “Role of red blood cells in haemostasis and thrombosis.” ISBT Sci Ser. Feb. 2017, 12(1):176-183.
Liu, X.H., et al., European Journal of Cancer, vol. 31A, No.6, pp. 953-963, (1995).
Llauger et al., “Evaluation of 8-Arylsulfanyl, 8-Arylsulfoxyl, and 8-Arylsulfonyl Adenine Derivatives as Inhibitors of the Heat Shock Protein 90”, J. Med. Chem., 48 (8), pp. 2892-2905, (Mar. 25, 2005).
Llauger et al., “Synthesis of 8-arylsulfoxyl/sulfonyl adenines”, Tetrahedron Letters, vol. 45, Issue 52, (Dec. 20, 2004), pp. 9549-9552.
Lochmatter, C. et al., Integrative phosphoproteomics links IL-23R signalling with metabolic adaption in lymphocytes, Scientific Reports, 6:24491 (2016).
Lockwood, S. et al., Endothelium-derived nitric oxide production is increased by ATP released from red blood cells incubated with hydroxyurea, Nitric Oxide, 38:1-7 (2014).
Loriga G. et al., Synthesis of 3,6-diazabicyclo [3.1.1]heptanes as novel ligands for the opioid receptors, Bioorganic & Medicinal Chemistry, Pergamon, GB, vol. 14, No. 3, pp. 676-691, (Feb. 1, 2006).
Lu, L., et al., “The HECT Type Ubiquitin Ligase NEDL2 is Degraded by Anaphase-promoting Complex/Cyclosome (APC/C)-Cdh1, and Its Tight Regulation Maintains the Metaphase to Anaphase Transition”, The Journal of Biological Chemistry, vol. 288, No. 50, pp. 35637-35650; (Dec. 2013).
Lucas, et al., “Facile Synthesis of a Library of 9-Alkyl-8-benzyl-9H-purin-6-ylamine Derivatives”, J. Comb. Chem., 3 (6), pp. 518-520, (Sep. 21, 2001).
MacDonald, Gregor J., et al, “Design and Synthesis of trans-3-(2-(4-((3-(3-(5-Methyl-1,2,4-oxadiazolyl))-phenyl(carboxamido)cyclohexy1)ethyl)-7-methylsulfonyl-2,3,4,5-tetrahydro-1H-3-benzazepine (SB-414796): A Potent and Selective Dopamine D3 Receptor Antagonist”, Journal of Medicinal Chemistry, 46(23), pp. 4952-4964 (2003).
Macdonald, Rosemary, Red cell 2,3-diphosphoglycerate and oxygen affinity, Anaesthesia, vol. 32, pp. 544-553, (1977).
Martinez-Mayorga Karina et al, Ligand/kappa-opioid receptor interactions: Insights from the X-ray crystal structure, European Journal of Medicinal Chemistry, vol. 66, pp. 114-121 (May 30, 2013).
Mathe-Allainmat, Monique et al., “Synthesis of 2-Amido-2, 3-dihydro-1H-phanalene Derivatives as New Conformationally Restricted Ligands for Melatonin Receptors”, Journal of Medicinal Chemistry, 39(16), pp. 3089-3095, (1996).
McCluskey A., et al., BioOrganic & Medicinal Chemistry Letters 10 (2000), pp. 1687-1690.
McCluskey A., et al., Bioorganic & Medicinal Chemistry Letters 11 (2001), pp. 2941-2946.
McGarry, E., et al., “The deubiquitinase USP9X maintains DNA replication fork stability and DNA damage checkpoint responses by regulating CLASPIN during S-phase”, Cancerres.aacrjournals.org, pp. 1-39; (2016).
Metcalf B, Chuang C, Dufu K, et al. “Discovery of GBT440, an orally bioavailable R-state stabilizer of sickle cell hemoglobin.” ACS Med Chem Lett. 2017; 8(3):321-326.
Meza, N.W. et al, In vitro and in vivo expression of human erythrocyte pyruvate kinase in erythroid cells: a gene therapy approach, Hum Gene Ther, 18(6):502-514 (2007).
Middelkoop, E. et al., Studies on sickled erythrocytes provide evidence that the asymmetric distribution of phosphatidylserine in the red cell membrane is maintained by both ATP-dependent translocation and interaction with membrane skeletal proteins, Biochimica et Biophysica Acta, 937:281-288 (1988).
Misra H. Bainbridge J, Berryman J, Abuchowski A, Galvez KM, Uribe LF, Hernandez AL, Sosa NR. “A phase 1b open label, randomized, safety study of SANGUINATE™ in patients with sickle cell anemia.” Rev Bras Hematol Hemoter. Jan.-Mar. 2017, 39(1):20-7.
Miwa, S. and Fujii, H., Molecular basis of erythroenzymopathies associated with hereditary hemolytic anemia: tabulation of mutant enzymes, Am J Hematol., 51(2): 122-132 (1996).
Moehrle, H., et al., “1,2,3,4-Tetrahydroquinolines as substrates for Mannich compounds”, Chemical Sciences, 53(7), pp. 742-752; (1998).
Moriyama R, Lombardo CR, Workman RF, Low PS. “Regulation of linkages between the erythrocyte membrane and its skeleton by 2,3-diphosphoglycerate.” J Biol Chem. May 25, 1993, 268(15):10990-6.
Mouchantaf, R., et al., “The Ubiquitin Ligase Itch is Auto-ubiquitylated in Vivo and in Vitro but is Protected from Degradation by Interacting with the Deubiquitylating Enzyme FAM/USP9X”, The Journal of Biological Chemistry, vol. 281, No. 50, pp. 38738-38747, (Dec. 2006).
Murn, J. et al., “Control of a neuronal morphology program by an RNA-binding zinc finger protein, Unkempt”, Genes & Development 29, pp. 501-512, (2015).
Murray, R.Z., Jolly, L.A., Wood, S.A., “The FAM Deubiquitylating Enzyme Localizes to Multiple Points of Protein Trafficking in Epithelia, where it Associates with E-cadherin and β-catenin”, Molecular Biology of the Cell, vol. 15, pp. 1591-1599; (Apr. 2004).
Muzyamba, M. and Gibson, J., Effect of 1-chloro-2,4-dinitrobenzene on K+ transport in normal and sickle human red blood cells, Journal of Physiology, 547(3):903-911 (2003).
Nagai, H., et al., “Ubiquitine-like Sequence in ASK1 Plays Critical Roles in the Recognition and Stabilization by USP9X and Oxidative Stress-Induced Cell Death”, Molecular Cell 36, pp. 805-818, (Dec. 2009).
Nagy, Peter I., et al., “Theoretical and Experimental Study on Ion-Pair Formation and Partitioning of Organic Salts in Octanol/Water and Dichloromethane/Water Sytems”, Journal of the American Chemical Society, 122 (28), pp. 6583-6593 (2000).
Nam, Keun-Soo et al., “Synthesis of quinolone antimicrobial agents and their antibacterial activities,” 5 Korean J. Med. Chem. (1995), pp. 2-5.
Narayanan, N., Wang, Z., Li, L., and Yang, Y., “Arginine methylation of USP9X promotes its interaction with TDRD3 and its anti-apoptotic activities in breast cancer cells”, Cell Discovery 3, pp. 1-17, (2017).
Nathan, J.A., et al., “The Ubiquitin E3 Ligase MARCH7 is Differentially Regulated by the Deubiquitylating Enzymes USP7 and USP9X”, Traffic, 9, pp. 1130-1145, (2008).
Neto, E.D. et al., “Shotgun sequencing of the human transcriptome with ORF expressed sequence tags”, PNAS, vol. 97, No. 7, pp. 3491-3496, (Mar. 2000).
Noma, T., et al., “Stage- and sex-dependent expressions of Usp9x, an X-linked mouse ortholog of Drosophila Fat facets, during gonadal development and oogenesis in mice”, Gene Expression Patters 2, pp. 87-91, (2002).
O'Connor, H.F., et al., “Ubiquitin-Activated Interaction Traps (UBAITs) identify E3 ligase binding partners”, EMBO reports, vol. 16, No. 12., (2015).
Obach RS. “Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes.” Drug Metab Dispos. Nov. 1999, 27(11):1350-9.
Oksenberg D, Dufu K, Patel MP, Chuang C, Li Z, Xu Q, Silva-Garcia A, Zhou C, Hutchaleelaha A, Patskovska L, Patskovsky Y, Almo SC, Sinha U, Metcalf BW, Archer DR. “GBT440 increases haemoglobin oxygen affinity, reduces sickling and prolongs RBC half-life in a murine model of sickle cell disease.” Br J Haematol. Oct. 2016, 175(1):141-53.
Oliviero, G., et al., “The variant Polycomb Repressor Complex 1 component PCGF1 interacts with a pluripotency sub-network that includes DPPA4, a regulator of embryogenesis”, pp. 1-11, (2015).
Olsen, J.V., et al., “Global, In Vivo, and Site-Specific Phosphorylation Dynamics in Signaling Networks”, Cell 127, pp. 635-648, (Nov. 2006).
Oski, M.D., Frank A., “The Role of Organic Phosphates in Erythrocytes on the Oxygen Dissociation of Hemoglobin,” Annals of Clinical Laboratory Science, vol. 1, No. 2 (Nov. 1970), pp. 162-176.
Ould Amar, A.K. et al., Assessment of qualitative functional parameters of stored red blood cells from donors with sickle cell trait (AS) or with heterozygote (AC) status, Transfus Clin Biol., 3(4):225-233 (1996).
Ouyang, W., et al., “β-catenin is regulated by USP9x and mediates resistance to TRAIL-induced apoptosis in breast cancer”, Oncology Reports 35, pp. 717-724, (2016).
OXBRYTA [package insert]. San Francisco, California, Global Blood Therapeutics, Inc. (Nov. 2019), 15 pgs.
OXBRYTA Slide Show—Jan. 2020.
Paemka, L., et al., “Seizures are Regulated by Ubiquitin-specific Peptidase 9 X-linked (USP9X), a De-Ubiquitinase”, PLOS Genetics, 11(3): pp. 1-16, (Mar. 2015).
Palsson-Mcdermott, EM et al., Pyruvate kinase M2 regulates Hif-1a activity and IL-1ß induction and is a critical determinant of the Warburg Effect in LPS-activated macrophages, Cell Metabolism, 21:65-80 (2015).
Papp, S.J., et al., “DNA damage shifts circadian clock time via Hausp-dependent Cry1 stabilization”, eLIFE, pp. 1-19, (2015).
Park, Yoon, Jin, Hyung-seung, and Liu, Yun-Cai, “Regulation of T cell function by the ubiquitin-specific protease USP9X via modulating the Carma 1-Bcl10-Malt1 complex”, PNAS, vol. 110, No. 23, pp. 9433-9438, (Jun. 2013).
Pászty C. “Transgenic and gene knock-out mouse models of sickle cell anemia and the thalassemias.” Curr Opin Hematol. 1997, 4(2):88-93.
Patel, P., et al., Synthesis of some novel pyrazoline and cyanopyridine derivatives as antimicrobial agents, Il Farmaco, vol. 51, No. 1, (1996), Abstract only.
Pavagadhi, T.H., et al., 3-(3′-phenoxyphenylmethyl)-5-aryl-1-acetylpyrazolines, Journal of the Institution of Chemists (India), vol. 73, No. 3, (2001), Abstract only.
Peddaboina, C. et al., “The downregulation of Mcl-1 via USP9X inhibition sensitizes solid tumors to Bcl-xl inhibition”, BMC Cancer, 12:541, pp. 1-12, (2012).
Perez-Mancera, P.A., et al., “The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma”, Nature, 486(7402): pp. 266-270; (Dec. 2012).
Platt OS. “Hydroxyurea for the treatment of sickle cell anemia.” N Engl J Med. 2008;358(13):1362-9.
Poillon W., & Kim, B., 2,3-Diphosphoglycerate and intracellular pH as interdependent determinants of the physiologic solubility of deoxyhemoglobin S, Blood, 76:1028-1036 (1990).
Poillon, W. et al., Antisickling effects of 2,3-Diphosphoglycerate Depletion, Blood, 85(11):3289-3296 (1995).
Poillon, W. et al., Intracellular hemoglobin S polymerization and the clinical severity of sickle cell anemia, Blood, 91:1777-1783 (1998).
Poillon, W. et al., The Effect of 2,3-Diphosphoglycerate on the Solubility of Deoxyhemoglobin S1, Archives of Biochemistry and Biophysics, vol. 249, No. 2, pp. 301-305, (Sep. 1986).
Press Release—“Agios Announces New Data from AG-348 and AG-519 Demonstrating Potential for First Disease-modifying Treatment for Patients with PK Deficiency” Dec. 4 2016—Globe Newswire
Press Release—“Agios Presents Updated Data from DRIVE PK Study Demonstrating AG-348 is Well-Tolerated and Results in Clinically Relevant, Rapid and Sustained Hemoglobin Increases in Patients with Pyruvate Kinase Deficiency” Dec. 10, 2017—Globe Newswire
PubChem SID: 440235168, modify date Feb. 25, 2021 (Feb. 25, 2021), Version 2, p. 1-7, Structure.
PubChem SID: 440235168, date Feb. 18, 2021 (Feb. 18, 2021), Version 1 of 2, p. 1-7, Structure.
PubChem CID: 135338361, create date: Dec. 15, 2018 (Dec. 15, 2018), p. 1, formula.
PubChem CID: 135338378, create date: Dec. 15, 2018 (Dec. 15, 2018), pg. 1, formula.
PubChem CID: 69203074, create date: Nov. 30, 2012 (Nov. 30, 2012), pp. 1-20, compound summary.
PubChem CID: 69203505, create date: Nov. 30, 2012 (Nov. 30, 2012), pp. 1-20, compound summary.
Rab, et al., AG-348 (Mitapivat), an allosteric activator of red blood cell pyruvate kinase, increases enzymatic activity, protein stability, and ATP levels over a broad range of PKLR genotypes, Haematologica, 1052xxx, (Jan. 23, 2020).
Rab, M.A.E. et al., Rapid and reproducible characterization of sickling during automated deoxygenation in sickle cell disease patients, Am. J. Hematol. (2019; 94; pp. 575-584.
Rabai M, Detterich JA, Wenby RB, et al. “Deformability analysis of sickle blood using ektacytometry.” Biorheology. 2014;51(2-3):159-70.
Ramdani, G. and Langsley, G., ATP, an Extracellular Signaling Molecule in Red Blood Cells: A Messenger for Malaria?, Biomed Journal, 37(5):284-292 (2014).
Raththagala, M. et al., Hydroxyurea stimulates the release of ATP from rabbit erythrocytes through an increase in calcium and nitric oxide production, European Journal of Pharmacology, 645(1-3):32-38 (2010).
REBLOZYL [package insert]. Cambridge, Massachusetts, Acceleron Pharma, Inc. (2020), 27 pgs.
REBLOZYL [package insert]. Summit, New Jersey, Celgene Corporation (Nov. 2019), 16 pgs.
Rice-Evans C, Omorphos SC, Baysal E. “Cell membranes and oxidative damage.” Biochem J. Jul. 1, 1986, 237(1):265-9.
Rosa, M. et al., Allosteric properties of hemoglobin and the plasma membrane of the erythrocyte: New insights in gas transport and metabolic modulation, Life, 60(2):87-93 (2008).
Ross, M.T., et al., “The DNA sequence of the human X chromosome”, Nature, 434, pp. 325-337; (Mar. 2005).
Rott, Ruth, et al., “α-Synuclein fate is determined by USP9X-regulated monoubiquitination”, PNAS, (2011).
Roy, R., et al., “HnRNPA1 couples nuclear export and translation of specific mRNAs downstream of FGF-2/S6K2 signalling”, Nucleic Acids Research, vol. 42, No. 20, pp. 12483-12497, (Oct. 2014).
Rush, J., et al., “Immunoaffinity profiling of tyrosine phosphorylation in cancer cells”, Nature Biotechnology, Vvol. 23, No. 1, pp. 94-101, (2005).
Sampson M, Archibong AE, Powell A, et al. “Perturbation of the developmental potential of preimplantation mouse embryos by hydroxyurea.” Int J Environ Res Public Health. 2010;7(5):2033-44.
Sato, Y., et al., “Ubiquitin-specific protease 9X in host cells interacts with herpes simplex virus 1 ICP0”, J. Vet. Med. Sci. 78(3), pp. 405-410; (2016).
Savio et al., “USP9X Controls EGFR Fate by Deubiquitinating the Endocytic Adaptor Eps15”, Current Biology 26, pp. 173-183, (Jan. 2016).
Schwartz, R. et al., Two distinct pathways mediate the formation of intermediate density cells and hyperdense cells from normal density sickle red blood cells, Blood, 92(12):4844-4855 (1998).
Schwickart, M., et al., “Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival”, Nature vol. 463, pp. 103-108; (Jan. 2010).
Sega, M. et al., Fluorescence assay of the interaction between hemoglobin and the cytoplasmic domain of erythrocyte membrane band 3, Blood Cells Mol Dis., 55(3):266-271 (2015).
Shen, G., et al., “MicroRNA-26b inhibits epithelial-mesenchymal transition in hepatocellular carcinoma by targeting USP9X,” BMC Cancer 14:393, pp. 1-11, (2014).
Shrestha, Archana, et al., “Oral Administration of FT-4202, an Allosteric Activator of Pyruvate Kinase-R, Has Potent Anti-Sickling Effects in a Sickle Cell Anemia (SCA) Mouse Model, Resulting in Improved RBC Survival and Hemoglobin Levels,” Blood (2020) 136 (Supplement 1):21-22, Nov. 4, 2020.
Shrestha, Archana, et al., “Oral Administration of FT-4202, an Allosteric Activator of Pyruvate Kinase-R, Has Potent Anti-Sickling Effects in a Sickle Cell Anemia (SCA) Mouse Model, Resulting in Improved RBC Survival and Hemoglobin Levels,” Presented at the 62nd American Society of Hematology (ASH) Annual Meeting, Dec. 5, 2020.
SIKLOS [package insert]. Lannoy, France, Delpharm Lille, (May 2019), 24 pgs.
SIKLOS [package insert]. Paris, France, Addmedica, (Dec. 2017), 25 pgs.
SIKLOS [package insert]. Paris, France, Addmedica, (May 2018), 23 pgs.
Smidrkal, Jan., “Synthesis of fagaronine”, Collection of Czechoslovak Chemical Communications, 53(12), pp. 3184-3192, (1988).
Sorathiya, S.D., et al., Preparation and antimicrobial activity of 3-(p-(2′,5′-dibromobenzenesulfonamido)phenyl)-5-aryl-1H/acetyl/phenyl-2-pyrazolines, Indian Journal of Chemistry, Section B: Organic, Incl. Medicinal Chemistry, vol. 36B, No. 7, (1997), Abstract only.
Soupene, E. and Kuypers, F., Identification of an erythroid ATP-dependent aminophospholipid transporter, British Journal of Haematology, 133(4):436-438 (2006)
Space SL, Lane PA, Pickett CK, Weil JV. “Nitric oxide attenuates normal and sickle red blood cell adherence to pulmonary endothelium.” Am J Hematol. Apr. 2000, 63(4):200-4.
Spinella, J.F., et al., “Genomic characterization of pediatric T-cell acute lymphoblastic leukemia reveals novel recurrent driver mutations”, Oncotarget, vol. 7, No. 40, pp. 65485-65503, (Sep. 2016).
Stasiuk, M. et al., Transformations of erythrocytes shape and its regulation, Postepy Biochem., 55(4):425-33 (2009). English Abstract.
St-Denis, N., et al., “Phenotypic and Interaction Profiling of the Human Phosphatases Identifies Diverse Mitotic Regulators”, Cell Reports 17, pp. 2488-2501, (Nov. 2016).
Stebbins et al., Crystal Structure of an Hsp90-Geldanamycin Complex: Targeting of a Protein Chaperone by an Antitumor Agent, Cell, (Apr. 1997), 89, p. 241.
Steinberg, Martin H., Pathophysiologically based drug treatment of sickle cell disease, TRENDS in Pharmacological Sciences, vol. 27, No. 4, (Apr. 2006).
Strausberg, R.L., et al., “Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences”, PNAS vol. 99, No. 26, pp. 16899-16903, (Dec. 2002).
Sun, H., et al., “Bcr-Abl ubiquitination and Usp9x inhibition block kinase signaling and promote CML cell apoptosis”, Blood, (Jan. 2011).
Sundd, Prithu et al., Pathophysiology of Sickle Cell Disease, Annual Review of Pathology: Mechanisms of Disease, (Oct. 9, 2018), pp. 261-290.
Swanson, Devin M. et al., “Identification and biological evaluation of 4-(3-trifluoromethylpyridine-2-yl) piperazine-1-c arboxylic acid (5-trifluoromethylpyridin-2-yl) amide, a high affinity TRPV1 (VR1) vanilloid receptor antagonist”, Journal of Medicinal Chemistry, 48(6), pp. 1857-1872 (2005).
Taipale, M., et al., “A Quantitative Chaperone Interaction Network Reveals the Architecture of Cellular Protein Homeostasis Pathways”, Cell 158, pp. 434-448, (Jul. 2014).
Takenaka, M. et al, Isolation and characterization of the human pyruvate kinase M gene, Eur J Biochem, 198(1):101-106 (1991).
Talmud, P.J., et al., “Gene-centric Association Signals for Lipids and Apolipoproteins Identified via the Human CVD Bead Chip”, The American Journal of Human Genetics 85, pp. 628-642, (Nov. 2009).
Tanphaichitr, V.S. et al, Successful bone marrow transplantation in a child with red blood cell pyruvate kinase deficiency, Bone Marrow Transplant, 26(6):689-690 (2000).
Taya, S., et al., “The deubiquitinating enzyme Fam interacts with and stabilizes ß-catenin”, Genes to Cells 4, pp. 757-767, (1999).
Taya, S., et al., “The Ras Target AF-6 is a Substrate of the Fam Deubiquitinating Enzyme”, The Journal of Cell Biology, vol. 142, No. 4, pp. 1053-1062, (Aug. 1998).
Telen, Marilyn, Malik, Punam, and Vercellotti, Gregory M., Therapeutic strategies for sickle cell disease: towards a multi-agent approach, Nature Reviews/Drug Discovery; (Dec. 4, 2018).
Terao, Y., et al., “Trifluoroacetic Acid-Catalyzed 1,3-Cycloaddition of the Simplest Iminium Ylide Leading to 3- or 3,4-Substituted Pyrrolidines and 2,5-Dihydropyrroles”, Chem. Pharm. Bull., 33(7), pp. 2762-2766, (1985).
Théard, D., et al., “USP9x-mediated deubiquitination of EFA6 regulates de novo tight junction assembly”, The EMBO Journal, vol. 29, No. 9, pp. 1499-1509, (2010).
Thein, Swee Lay, The Molecular Basis of Bß-Thalassemia, Cold Spring Harb Perspect Med. (2013).
Thompson, Alexis, M.D., M.P.H., “A Targeted Agent for Sickle Cell Disease—Changing the Protein but Not the Gene,” The New England Journal of Medicine, (Jun. 14, 2019).
Tian, S., et al., Yaoxue Xueba (1993), 28(11), pp. 870-875. Chemical Abstract No. 120:299229.
Toloczko, A., et al., “Deubiquitinating Enzyme USP9X Suppresses Tumor Growth via LATS kinase and Core Components of the Hippo pathway”, Cancer Research, pp. 1-37, (Jul. 2017).
Tripathi, Ashutoshi and Safo, Martin K., In Silico-Screening Approaches for Lead Generation: Identification of Novel Allosteric Modulators of Human-Erythrocyte Pyruvate Kinase, Allostery: Methods and Protocols, Methods in Molecular Biology, Chpt. 19, vol. 796, pp. 351-367 (2012).
Trivigno, D., et al., “Deubiquitinase USP9x Confers Radioresistance through Stabilization of Mcl-1 1,2”, NEO Plasia, vol. 14, No. 10, pp. 893-904, (Oct. 2012).
Tsai, Y.C., et al., “Functional Proteomics Establishes the Interaction of SIRT7 with Chromatin Remodeling Complexes and Expands Its Role in Regulation of RNA Polymerase I Transcription”, Molecular & Cellular Proteomics 11.5, pp. 60-76, (2012).
Tsutsumi H, Tani K, Fujii H, Miwa S. “Expression of L- and M-type pyruvate kinase in human tissues. Genomics.” 1988, 2(1):86-9.
United States Securities and Exchange Commission, Form S-1 Registration Statement, Forma Therapeutics Holdings, Inc., dated Dec. 8, 2020, 374 pages.
United States Securities and Exchange Commission, Form S-1, Registration Statement—Forma Therapeutics Holdings, Inc., May 29, 2020.
Upadhyay J., et al., Studies on pyrazolines. Part III. Preparation and antimicrobial activity of 3-(4-phenylsulfonamidophenyl)-5-aryl-1-ace tyl/phenyl-4,5-dihydropyrazoles, Journal of the Indian Chemical Society, vol. 68, No. 7, (1991), pp. 413-414.
Van Zweiten, R. et al., Inborn defects in the antioxidant systems of human red blood cells, Free Radio Biol Med., 67:377-386 (2014).
Vanderah et al, Novel d-amino acid tetrapeptides produce potent antinociception by selectively acting at peripheral kappa-opioid receptors, European Journal of Pharmacology, Elsevier Science, vol. 583, No. 1, pp. 62-72 (Jan. 24, 2008).
Varjosalo, M., et al., The Protein Interaction Landscape of the Human CMGC Kinase Group, Cell Reports 3, pp. 1306-1320, (Apr. 2013).
Verma, S.K. et al., Imidazole-Catalyzed Monoacylation of Symmetrical Diamines, Organic Letters, 12(19): 4232-4235 (2010).
Vichinsky, E. et al., “A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease,” N Engl J Med. DOI: 10.1056/NEJMoa1903212 (Jun. 2019).
Vichinsky, E. et al., Protocol to A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease, (Jun. 14, 2019).
Vichinsky, E. et al., Supplementary Appendix to A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease, (Jun. 14, 2019).
Vong, Q. P., et al., “Chromosome Alignment and Segregation Regulated by Ubiquitination of Survivin”, Science, vol. 310, pp. 1499-1504, (Dec. 2, 2005).
Voskou S, Aslan M, Fanis P, Phylactides M, Kleanthous M. “Oxidative stress in β-thalassaemia and sickle cell disease.” Redox Biol. Dec. 2015, 6:226-39.
Wagner, G. et al., Red cell vesiculation—a common membrane physiologic event, J Lab Clin., 108(4):315-324 (1986).
Wan, C., et al., “Panorama of ancient metazoan macromolecular complexes”, Nature 525(7569), pp. 339-344, (Sep. 2015).
Wang, G.S., et al., Journal of Ethnopharmacology, 26 (1989), pp. 147-162.
Wang, H. et al., JMJD5 regulates PKM2 nuclear translocation and reprograms HIF-1a-mediated glucose metabolism, PNAS, 111(1):279-284 (2014).
Wang, J., et al, “TopBP1 Controls BLM Protein Level to Maintain Genome Stability”, Molecular Cell 52, pp. 667-678, (Dec. 2013).
Wang, Q., et al., “The X-linked Deubiquitinase USP9X is an Integral Component of Centrosome”, The American Society for Biochemistry and Molecular Biology, Inc., pp. 1-33, (2017).
Wang, S. et al., “Ablation of the oncogenic transcription factor ERG by deubiquitinase inhibition in prostate cancer”, PNAS, vol. 111, No. 11, pp. 4251-4256, (Mar. 2014).
Wang, S., et al., “The ubiquitin ligase TRIM25 targets ERG for degradation in prostate cancer”, Oncotarget, vol. 7, No. 40, pp. 64921-64931, (2016).
Wang, X, et al., “Hsp90 Cochaperone Aha1 Downregulation Rescues Misfolding of CFTR in Cystic Fibrosis”, Cell 127, pp. 803-815, (Nov. 2006).
Waza et al., Nature, 11, No. 10, (Oct. 2005), pp. 1088-1095.
Weatherall, D., The inherited diseases of hemoglobin are an emerging global health burden, Blood, 115(22):4331-43336 (2010).
Wei, Wan-Guo et al., “A practical procedure for multisubstituted .beta.-naphthols and their derivatives”, Tetrahedron, 59(34), pp. 6621-6625, (2003).
Willcocks, J. et al., Simultaneous determination of low free Mg2+ and pH in human sickle cells using P NMR spectroscopy, The Journal of Biological Chemistry, 277(51):49911-49920 (2002).
Wood BL, Gibson DF, Tait JF. “Increased erythrocyte phosphatidylserine exposure in sickle cell disease: flow-cytometric measurement and clinical associations.” Blood., 88(5):1873-80 (Sep. 1, 1996).
Wood, Kenneth W., et al., “An Adaptive, Randomized, Placebo-Controlled, Double-Blind, Multi-Center Study of Oral FT-4202, a Pyruvate Kinase Activator in Patients with Sickle Cell Disease (PRAISE),” Blood (2020) 136 (Supplement 1):19-20, Nov. 4, 2020.
Wood, Kenneth W., et al., “An Adaptive, Randomized, Placebo-Controlled, Double-Blind, Multi-Center Study of Oral FT-4202, a Pyruvate Kinase Activator in Patients with Sickle Cell Disease,” Presented at the 62nd American Society of Hematology (ASH) Annual Meeting, Dec. 7, 2020.
Woods, N.T., et al., “Charting the Landscape of Tandem BRCT Domain-Mediated Protein Interactions”, Sci Signal, 5(242), pp. 1-35, (2014).
Wright, S.W. et al., A Convenient Preparation of Heteroaryl Sulfonamides and Sulfonyl Fluorides from Heteroaryl Thiols, J. Org. Chem., 71: 1080-1084 (2006).
Wu, Y., et al., “Aberrant phosphorylation of SMAD4 Thr277-mediated USP9x-SMAD4 interaction by free fatty acids promotes breast cancer matastasis”, Cancer Research, pp. 1-34, (2017).
Wu, Z., et al., “Targeted Ubiquitination and Degradation of G-Protein-Coupled Receptor Kinase 5 by the DDB1-CUL4 Ubiquitin Ligase Complex”, PLOS One, vol. 7, Issue 8, pp. 1-11, (Aug. 2012).
Xie, Y., et al., “Deubiquitinase FAM/USP9X Interacts with the E3 Ubiquitin Ligase SMURF1 Protein and Protects it from Ligase Activity-dependent Self-degradation”, The Journal of Biological Chemistry., vol. 288, No. 5, pp. 2976-2985, (Feb. 2013).
Xu, Z., et al., “Identification of a Deubiquitinating Enzyme as a Novel AGS3-Interacting Protein”, PLOS One, vol. 5, Issue 3, pp. 1-12, (Mar. 2010).
Yan, J., et al., “Usp9x- and Noxa-mediated Mcl-1 downregulation contributes to pemetrexed-induced apoptosis in human non-small-cell lung cancer cells”, Cell Death and Disease 5, pp. 1-7, (2014).
Yang H, Merica E, Chen Y, Cohen M, Goldwater R, Hill C, et al. “Phase I single (SAD) and multiple ascending dose (MAD) studies of the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of AG-348, a first-in-class allosteric activator of pyruvate kinase-R, in healthy subjects.” Blood. 2014, 124:4007.
Yang H, Merica E, Chen Y, et al. “Phase 1 Single- and Multiple-Ascending-Dose Randomized Studies of the Safety, Pharmacokinetics, and Pharmacodynamics of AG-348, a First-in-Class Allosteric Activator of Pyruvate Kinase R, in Healthy Volunteers.” Clin Pharmacol Drug Dev. Aug. 9, 2018.
Yang, H. et al., Phase 1 Single- and Multiple-Ascending-Dose Randomized Studies of the Safety, Pharmacokinetics, and Pharmacodynamics of AG-348, a First-in-Class Allosteric Activator of Pyruvate Kinase R, in Healthy Volunteers, 8 Clin. Pharmacol. Drug Dev. 246-259 (2019)
Yi, S., et al., Leukemia Research, vol. 15(10), (1991), pp. 883-886.
You, J. and Pickart, C.M., “A HECT Domain E3 Enzyme Assembles Novel Polyubiquitin Chains”, vol. 276, No. 23, pp. 19871-19878, (2001).
Yu, W., et al., “Large-Scale Concatenation cDNA Sequencing”, Genome Research 7, pp. 353-358, (1997).
Zanella A, Fermo E, Bianchi P, Chiarelli LR, Valentini G. “Pyruvate kinase deficiency: The genotype-phenotype association.” Blood Rev. 2007, 23:217-31.
Zanella A, Fermo E, Bianchi P, Valentini G. “Red cell pyruvate kinase deficiency: molecular and clinical aspects.” Br J Haematol. 2005;130(1):11-25.
Zhang, C., et al., “Synergistic anti-tumor activity of gemcitabine and ABT-737 in vitro and in vivo through disrupting the interaction of USP9X and Mcl-1”, Molecular Cancer Therapeutics, (May 12, 2011).
Zhang, C., et al., “USP9X destabilizes pVHL and promotes cell proliferation”, Oncotarget, vol. 7, No. 37, pp. 60519-60534, (2016).
Zhang, Y & Xia, Y., Adenosine signaling in normal and sickle erythrocytes and beyond, Microbes Infect., 14(10) (2012).
Zhang, Y. et al., Detrimental effects of adenosine signaling in sickle cell disease, Nature Medicine, 17(1):79-87 (2011).
Zhang, Yongmin et al., “Organic reactions in chiral micelles. 7. The structural effects on the asymmetric oxidation of prochiral sulfides in chiral micelles”, Chinese Journal of Chemistry, (1990), pp. 89-96.
Zhao, Y., et al., “Noncanonical regulation of alkylation damage resistance by the OTUD4 deubiquitinase”, EMBO Journal, vol. 34, No. 12, pp. 1687-1703, (2015).
Zhi et al., Hybrid Antibacterals. DNA Polymerase—Topoisomerase Inhibitors. J. Med. Chem., published on Web Jan. 25, 2006., vol. 49, pp. 1455-1465, especially p. 1456. Scheme 3, compound 4; p. 1457, Scheme 4, compound 13, p. 1462.
Zhou, L., et al., “The Scaffold Protein KSR1, A Novel Therapeutic Target for the Treatment of Merlin-Deficient Tumors”, Oncogene 35(26), pp. 3443-3453, (Jun. 2016).
Zhou, ZH et al., Phosphorus, Sulfur and Silicon and the Related Elements (1999), 152, pp. 45-52. Chemical Abstract No. 132: 180853.
Zhu, Tong et al., Polymer-Supported Synthesis of Pyridone-Focused Libraries as Inhibitors of Anaplastic Lymphoma Kinase, Journal of Combinatorial Chemistry, 2006, 8(3), pp. 401-409.
National Center for Biotechnology Information. PubChem Substance Record for SID 377251214, SCHEMBL20511283, Source: SureChEMBL. https://pubchem.ncbi.nlm.nih.gov/substance/377251214. Accessed Nov. 3, 2020. Available Dec. 15, 2018. (Year: 2018).
U.S. Appl. No. 16/576,720, filed Sep. 19, 2019 , 47 pages.
Qian et al., “Drug-polymer solubility and miscibility: Stability consideration and practical challenges in amorphous solid dispersion development”, J. Pharm. Sci., Jul. 2010, vol. 99, No. 7, pp. 2941-2947.
Related Publications (1)
Number Date Country
20230381151 A1 Nov 2023 US
Provisional Applications (5)
Number Date Country
62811904 Feb 2019 US
62789641 Jan 2019 US
62782933 Dec 2018 US
62733558 Sep 2018 US
62733562 Sep 2018 US
Continuations (2)
Number Date Country
Parent 17008787 Sep 2020 US
Child 18087774 US
Parent 16576720 Sep 2019 US
Child 17008787 US