This disclosure relates to compositions and methods for the treatment of viral diseases, including COVID-19.
The coronavirus disease 2019 (COVID-19) pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has already infected hundreds of millions of people worldwide and is responsible for millions of deaths (Dong, E., et al. The Lancet. Infectious diseases 20, 533-534, doi:10.1016/S1473-3099(20)30120-1 (2020); Siddiqi, H. K. & Mehra, M. R. J Heart Lung Transplant 39, 405-407, (2020); Dolken, L., et al. Viruses 13, (2021)). A subset of patients develop a severe form of the disease which is associated with the presence of a large set of pro-inflammatory cytokines and an excessively exaggerated immune response, the so-called cytokine storm or hypercytokinemia, that are produced by macrophages in the lungs (Tay, M. Z., et al. Nat Rev Immunol 20, 363-374, (2020); Abassi, Z., et al. Front Immunol 11, 1312, (2020); Huang, C. et al. Lancet 395, 497-506, (2020); Melms, J. C. et al. Nature 595, 114-119, (2021); Rendeiro, A. F. et al. Nature 593, 564-569, (2021); Liao, M. et al. Nat Med, 26, 842-844 (2020); Hadjadj, J. et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718-724, (2020). How the anti-viral response to SARS-CoV-2 can evolve into a hyperactivation syndrome with the recruitment and subsequent activation of macrophages in the lungs is currently unclear. CS is also associated with selected viral infections, particularly influenza, SARS, and hantavirus, and often results in mortality and morbidity (Oldstone MB, Rosen H., Curr Top Microbiol Immunol. 2014; 378:129-47.)
Safe and effective drugs to control the cytokine storm are lacking, and clinically, the treatment of the cytokine storm has proved difficult (Yang, L., et al. Sig Transduct Target Ther 6, 255 (2021). Thus, there is a tremendous need for effective therapeutics to counter the cytokine storm associated with viral diseases.
This disclosure relates to methods for treating viral diseases associated with the cytokine storm in a human subject using a compound that that disrupts the tri-carboxylic acid (TCA) cycle and/or activates the Unfolded Protein response (UPR) in cells (e.g., dendritic cells and macrophages).
In the first aspect, the disclosure features a method of treating a condition selected from the group consisting of a viral disease or hypercytokinemia in a human subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound that disrupts the tri-carboxylic acid (TCA) cycle in immune cells or a compound that activates the Unfolded Protein response (UPR) in immune cells in the subject.
In some embodiments, the hypercytokinemia comprises an overproduction of immune cells and pro-inflammatory cytokines into the lungs of the subject.
In some embodiments, the compound that activates the UPR activates the IRE1α-XBP1 signaling branch of the UPR in immune cells.
In some embodiments, the compound that activates the UPR is tunicamycin, or thapsigargin. In some embodiments, the compound that activates the UPR is IXA4.
In some embodiments, the immune cells are dendritic cells, macrophages, T cells, B cells, natural killer cells, and/or neutrophils.
In some embodiments, the compound that disrupts the tri-carboxylic acid (TCA) cycle is
or a pharmaceutically acceptable salt thereof, wherein R1 and R2 are independently selected from the group consisting of acyl defined as R3C(O)—, alkyl defined as CnH2n+1, alkenyl defined as CmH2m−1, alkynyl defined as CmH2m−3, aryl, heteroaryl, alkyl sulfide defined as CH3(CH2)n—S—, imidoyl defined as R3C(═NH)—, hemiacetal defined as R4CH(OH)—S—, and hydrogen provided that at least one of R1 and R2 is not hydrogen; wherein R1 and R2 as defined above can be unsubstituted or substituted; wherein R3 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, or heterocyclyl, any of which can be substituted or unsubstituted; wherein R4 is CCl3 or COOH; and wherein x is 0-16, n is 0-10 and m is 2-10,
In some embodiments, the R1 and R2 are benzyl or benzoyl.
In some embodiments, the compound of Formula I is
In some embodiments, the compound of formula I is 6,8-bis-benzylthio-octanoic acid.
In some embodiments, the viral disease is coronavirus disease-19 (COVID-19), influenza, Severe Acute Respiratory Syndrome (SARS), or Hantavirus Pulmonary Syndrome (HPS).
In some embodiments, the subject is concurrently treated with one or more agents selected from the group consisting of a corticosteroid, remdesivir, Nirmatrelvir, Bebtelovimab, Molnupiravir, an IL-6 inhibitor, an IL-1 inhibitor, a kinase inhibitor, a complement inhibitor, ivermectin, hydroxychloroquine, favipiravir, interferon-beta and a nonsteroidal anti-inflammatory drug (NSAID). In some embodiments, the immunosuppressant is methotrexate, mycophenolate mofetil (MMF), cyclophosphamide, cyclosporin, or azathioprine. In some embodiments, the corticosteroid is hydrocortisone, methylprednisolone, dexamethasone or prednisone.
In some embodiments, the treatment reduces production of inflammatory cytokines or chemokines by dendritic cells in the human subject. In some embodiments, the inflammatory cytokines or chemokines are selected from the group consisting of: type I interferon (IFN-I), IL-6, or TNF-α, type III interferon, MIP-1a/CCL3, MIP-1/CCL4, CCL5/RANTES, and IP-10/CXCL10.
In some embodiments, the dendritic cells are plasmocytoid dendritic cells. In some embodiments, the dendritic cells express one or more of CD123, CD303 (BDCA2), CD304 (BDCA4), and immunoglobulin-like transcript 7 (ILT7). In some embodiments, the dendritic cells do not express the lineage-associated markers (Lin) CD3, CD19, CD14, CD16 and CD11c.
In some embodiments, the method inhibits and/or reduces IFN-I production in the human subject in need thereof by at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99%, as compared to the corresponding reference levels in the human subject or in a control.
In some embodiments, the treatment reduces the expression of one or more of the interferon stimulated genes selected from the group consisting of Guanylate Binding Protein 1 (GBP1), Interferon Regulatory Factor 7 (IRF7), interferon stimulated gene 54 (ISG54), myxovirus resistance protein B (MxB), and 2′-5′-Oligoadenylate Synthetase 2 (OAS2).
In some embodiments, the treatment enhances expression of phosphoglycerate dehydrogenase (PHGDH), phosphoserine Phosphatase (PSPH), and phosphoserine Aminotransferase 1 (PSAT1).
In another aspect, the disclosure features the use of a therapeutically effective amount of a compound that disrupts the tri-carboxylic acid (TCA) cycle or a compound activates the Unfolded Protein response (UPR) in immune cells in the subject to treat a condition selected from the group consisting of a viral disease and hypercytokinemia in the subject.
In some embodiments, the compound that disrupts the tri-carboxylic acid (TCA) cycle is (a) a compound of Formula I
or a pharmaceutically acceptable salt thereof, wherein R1 and R2 are independently selected from the group consisting of acyl defined as R3C(O)—, alkyl defined as CnH2n+1, alkenyl defined as CmH2m−1, alkynyl defined as CmH2m−3, aryl, heteroaryl, alkyl sulfide defined as CH3(CH2)n—S—, imidoyl defined as R3C(═NH)—, hemiacetal defined as R4CH(OH)—S—, and hydrogen provided that at least one of R1 and R2 is not hydrogen; wherein R1 and R2 as defined above can be unsubstituted or substituted; wherein R3 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylaryl, heteroaryl, or heterocyclyl, any of which can be substituted or unsubstituted; wherein R4 is CCl3 or COOH; and wherein x is 0-16, n is 0-10 and m is 2-10,
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
This disclosure is based, in part, on the findings that interferon-I is produced during the cytokine storm response by plasmacytoid dendritic cells (pDCs)s in SARS-CoV-2-infected patients. Further, the infiltration of pDCs in the lungs of SARS-CoV-2-infected patients correlated with strong IFN-I signaling in lung macrophages. In patients that developed severe COVID-19, lung macrophages primed by IFN-I express a robust inflammatory signature which correlated at the single cell level with persisting IFN-I signaling. Further, SARS-CoV-2-activated pDCs (SARS pDCs) induced IFN-α-mediated transcriptional and epigenetic mechanisms that lead to the hyperactivation of macrophages by environmental stimuli. Without being bound by theory, it is believed that the uncoupling of the kinetics of the IFN-I signature with the cytokine storm in lung macrophages indicates a link between pDC's response to SARS-CoV-2 and the subsequent macrophage activation in patients with severe COVID-19. Further, the data described points to pDCs, by direct sensing of SARS-CoV-2, and to monocyte-derived macrophages indirectly by phagocytosis of infected cells, as the likely source of IFN-I in the lungs of infected individuals. By promoting the macrophage-induced cytokine storm, this IFN-I response can lead to devastating consequences for a subset of patients.
The Inositol-Requiring Enzyme-X-Box Binding Protein 1 (IRE1α-XBP1 branch of the unfolded protein response (UPR)) inhibits the production of IFN-I by toll-like receptor (TLR)-activated plasmacytoid dendritic cells (pDCs). Mechanistically, IRE1α-XBP1 activation reprograms glycolysis to serine metabolism by inducing phosphoglycerate dehydrogenase (PHGDH) expression. This reduces pyruvate access into the tricarboxylic (TCA) cycle and blunts mitochondrial ATP generation that is necessary for IFN-I production. Furthermore, decreased expression of PHGDH and UPR-controlled genes in pDCs purified from patients with systemic sclerosis (SSc) was observed. Accordingly, pharmacological blockade of tri-carboxylic acid (TCA) cycle reactions can inhibit IFN-I responses in pDCs of patients. These findings link the UPR to metabolic control of pDC hyperactivation and suggest that modulating this process may represent an unconventional strategy for the treatment of viral diseases associated with the cytokine storm.
Thus, this disclosure features agents that activate the UPR response in immune cells such as dendritic cells, and agents that disrupt the TCA cycle in such cells. The disclosure features methods of using such agents to treat a human subject with a viral disease (e.g., COVID-19) and/or to reduce production of inflammatory cytokines or chemokines by immune cells such as DCs (e.g., type I interferon (IFN-I), IL-6, or TNF-α, type III interferon, MIP-1a/CCL3, MIP-1/CCL4, CCL5/RANTES, and IP-10/CXCL10).
A detailed description of the UPR activating agents and the TCA cycle disrupting agents, as well as methods of using these agents are set forth below.
The tricarboxylic acid (TCA) cycle (also called the Krebs cycle) is the second stage of cellular respiration. It is a series of chemical reactions to release stored energy through the oxidation of acetyl-CoA derived from carbohydrates, fats, and proteins. In eukaryotic cells, the citric acid cycle occurs in the matrix of the mitochondrion. In the context of this disclosure, the term “disrupt”, with respect to the TCA cycle disrupting agents refers to agents that inhibit mitochondrial metabolism in cells such as immune cells (macrophages and dendritic cells). The TCA cycle and exemplary inhibitors thereof are shown in
In some embodiments, the TCA cycle disrupting agent is any of the compounds of Formula I or a pharmaceutically acceptable salt thereof as described in U.S. Pat. No. 9,839,691, incorporated by reference in its entirety. A compound of Formula I has the following structure:
In some embodiments, the compound of formula I is 6,8-bis-benzylthio-octanoic acid (CPI-613 or Devimistat).
In some embodiments, the TCA cycle disrupting agent is UK5099 which inhibits mitochondrial pyruvate carrier, a carrier which transport pyruvate from cytoplasm to mitochondria. UK5099 has the following structure:
In some embodiments, the TCA cycle disrupting agent is CB839 (Telagenastat), which inhibits glutaminase, an enzyme that converts glutamine to glutamate. CB839 has the following structure:
The unfolded protein response (UPR) is an adaptive response that maintains the fidelity of the cellular proteome in conditions that subvert the folding capacity of the cell, such as those noticed in infection and inflammatory contexts. In immunity, the UPR sensor IRE1 (Inositol-requiring enzyme 1-alpha) is as a critical regulator of the homeostasis of antigen presenting cells (APCs). Flores-Santibinez F, et al. Cells. 2019; 8(12):1563. The IRE1α/XBP1s signaling pathway is an arm of the unfolded protein response (UPR) that safeguards the fidelity of the cellular proteome during endoplasmic reticulum (ER) stress, and that has also emerged as a key regulator of dendritic cell (DC) homeostasis. Medel B. et al., Frontiers in Immunology, 2019(9); Article 3050.
In the context of this disclosure, compounds that activate the UPR in plasmocytoid dendritic cells, particularly the IRE1α/XBP1s signaling pathway, can be used in the methods to treat viral diseases and/or to reduce proinflammatory cytokine production. Such UPR activating agents include, but are not limited to tunicamycin and thapsigargin. As described herein, the term “activates the UPR” refers to the ability of the agent to activate and/or enhance the unfolded protein response, in particular, the IRE1α-XBP1 signaling branch of the UPR in cells (e.g., immune cells such as macrophages and dendritic cells).
Exemplary UPR activating agents that can be utilized in the methods described herein have the structures provided below:
Any of the UPR activating agents shown in Table 1 or analogs thereof can be utilized in the methods of this disclosure.
The disclosure features methods of treating viral diseases, in particular viral diseases associated with the cytokine storm or hypercytokinemia. Cytokine storm defines a dysregulation of and an excessively exaggerated immune response often accompanying selected viral infections, such as influenza, SARS (including SARS-CoV-2), MERS, and hantavirus. Cytokine storm is an umbrella term encompassing several disorders of immune dysregulation characterized by constitutional symptoms, systemic inflammation, and multiorgan dysfunction that can lead to multiorgan failure if inadequately treated. See e.g., Fajgenbaum and June N Engl J Med 2020; 383.2255-2273.
Nonspecific markers of inflammation such as C-reactive protein (CRP) are universally elevated and correlate with severity. Other symptoms of a cytokine storm include, but are not limited to hypertriglyceridemia and various blood-count abnormalities, such as leukocytosis, leukopenia, anemia, thrombocytopenia, and elevated ferritin and d-dimer levels. Prominent elevations in serum inflammatory cytokine levels, including but not limited to interferon-γ(or CXCL9 and CXCL10, chemokines induced by interferon-γ), interleukin-6, interleukin-10, and soluble interleukin-2 receptor alpha, a marker of T-cell activation, are usually present in a cytokine storm. Innate cells that are most often implicated in the pathogenesis of cytokine storm include neutrophils, macrophages, and NIK cells.
Serum cytokine levels that are elevated in patients with Covid-19-associated cytokine storm include but are not limited to interleukin-1β, interleukin-6, IP-10, TNF, interferon-γ, macrophage inflammatory protein (MIP) 1α and 1β, and VEGF. In some embodiments, the disclosued methods of this disclosure treat the cytokine storm by reducing levels of these cytokines.
Apart from SARS-CoV-2 infections that cause coronavirus disease (COVID-19), the methods of this disclosure can be used to treat other viral diseases, including, but not limited to influenza Severe Acute Respiratory Syndrome (SARS), and Hantavirus Pulmonary Syndrome (HPS).
In some embodiments, the methods of the disclosure can treat viral conditions which are IFN-I-mediated. The methods of this disclosure may be used to block the TCA cycle and/or activate the UPR in a range of immune cells, including, but not limited to, dendritic cells, macrophages, T cells, B cells, natural killer cells, and/or neutrophils. Several types of immune cells that are involved in the pathology of viral diseases. See e.g., Tang L et al., Front. Immunol., Nov. 30, 2020.
The virus can promote the activation of immune cells (such as T cells, B cells, macrophages, dendritic cells, neutrophils, monocytes) and resident tissue cells, resulting in the production of large amounts of inflammatory cytokines. During the flu virus infection, innate immune responses get started through the cascade amplification reactions of interferon stimulated gene expression, and type I interferon (IFN) is mainly produced by monocytes, macrophages and dendritic cells.
Serum levels of interleukin 8 (IL-8), IP-10 (interferon-induced protein 10), MCP-1 (monocyte chemoattractant protein-1), MIP-1 (macrophage inflammatory protein-1), MIG (monokine induced by IFN-γ) and CXCL-9 (CXC chemokine ligand-9) can be abnormally elevated in H5N1 influenza virus infection, while IL-8, IL-9, IL-17, IL-6, IL-15, TNF-α (tumor necrosis factor-α), IL-10 can be increased in H1N1 influenza virus infection. Earlier researches demonstrated that serum levels of proinflammatory factors IFN-γ, IL1β, IL-6, IL-12, IL-18, IP-10, MCP-1, and CCL2 (CC chemokine ligand-2), CXCL-10 and IL-8 are positively correlated with lung inflammation and extensive lung tissue injury in SARS patients (19-21). Whereas, the levels of serum pro-inflammatory cytokines IL-6, IFN-γ, TNF-α, IL-15, IL-17, and chemokines IL-8, CXCL-10, and CCL5 were significantly increased in severe MERS patients (22, 23). Among numerous molecules that increase in virally-mediated cytokine storms, IL-6, IFN-γ, IL-1β, IL-8, IL-10, and TNF-α are of crucial importance (9, 24, 25). The occurrence of cytokine storm has been reported to be one of the main causes of death in patients with SARS-CoV, MERS-CoV, and influenza virus infections (8, 26). Similarly, cytokine storm is also a common feature of severe cases in COVID-19, and elevated levels of serum IL-6 and CRP correlate with respiratory failure, ARDS, MOF and adverse clinical outcomes (27, 28).
Plasmocytoid dendritic cells (pDCs) are danger-sensing cells that produce interferon (IFN)—I. IFNs are generally classified into three families—IFN-I, IFN-II and IFN-III—which differ in their immunomodulatory properties, their structural homology and the group of cells from which they are secreted [3, 4]. IFN-Is (IFN-α, -β, -ω, -ε, -κ) compose the largest family and, alongside IFN-III (IFN-λ), activates intracellular signaling pathways that mediate immune responses against viruses and tumors. (Psarras A et al., Rheumatology, Volume 56, Issue 10, October 2017, Pages 1662-1675).
pDCs play a crucial role in antiviral immunity and are regarded as precursor DC which are effectively interferon producing cells. In some embodiments, the methods of the disclosure can be used to modulate signaling pathways in pDCs and other immune cells, thereby treating viral conditions. In some embodiments, the pDCs that are modulated are dendritic cells express one or more of CD123, CD303 (BDCA2), CD304 (BDCA4), and immunoglobulin-like transcript 7 (ILT7), but do not express the lineage-associated markers (Lin) CD3, CD19, CD14, CD16 and CD11c. See, e.g., Ye, Y, et al., Clinical & Translational Immunology (2020); 9: e1139; Tang F, Du Q, Liu Y J. Sci China Life Sci. 2010 February; 53(2):172-82; Gilliet M, Cao W, Liu Y J. Nat Rev Immunol. 2008 August; 8(8):594-606; and Barrat F. J. and Su L., J Exp Med. 2019 Sep. 2; 216(9):1974-1985 for a review of pDCs, characterization of these cells, and their role in viral conditions.
This disclosure features combination therapies wherein the UPR activating agent and/or the TCA cycle disruptor is administered with one or more additional treatments. The additional treatment can be an art-recognized therapy for a viral diseases (e.g., COVID-19). See Kheirabadi D, et al., J Med Virol. 2021; 93(5):2705-2721. doi:10.1002/jmv.26811. Treatments for viral disease include, but are not limited to, a corticosteroid, remdesivir, Nirmatrelvir, Bebtelovimab, Molnupiravir, an IL-6 inhibitor, an IL-1 inhibitor, a kinase inhibitor, a complement inhibitor, ivermectin, hydroxychloroquine, favipiravir, interferon-beta and a nonsteroidal anti-inflammatory drug (NSAID). Immunosuppressant treatment includes, but is not limited to, treatment with methotrexate, mycophenolate mofetil (MMF), cyclophosphamide, cyclosporin, or azathioprine. The corticosteroid that can be used as an additional treatment includes, but is not limited to, dexamethasone, prednisone, hydroxyl-triamcinolone, alpha-methyl dexamethasone, dexamethasone-phosphate, beclomethasone dipropionates, clobetasol valerate, desonide, desoxymethasone, desoxycorticosterone acetate, dexamethasone, dichlorisone, diflorasone diacetate, diflucortolone valerate, fluadrenolone, fluclorolone acetonide, fludrocortisone, flumethasone pivalate, fluosinolone acetonide, fluocinonide, flucortine butylesters, fluocortolone, fluprednidene (fluprednylidene)acetate, flurandrenolone, halcinonide, hydrocortisone acetate, hydrocortisone butyrate, methylprednisolone, triamcinolone acetonide, cortisone, cortodoxone, flucetonide, fludrocortisone, difluorosone diacetate, fluradrenolone, fludrocortisone, diflurosone diacetate, fluradrenolone acetonide, medrysone, amcinafel, amcinafide, betamethasone and the balance of its esters, chloroprednisone, chlorprednisone acetate, clocortelone, clescinolone, dichlorisone, diflurprednate, flucloronide, flunisolide, fluoromethalone, fluperolone, fluprednisolone, hydrocortisone valerate, hydrocortisone cyclopentylpropionate, hydrocortamate, meprednisone, paramethasone, prednisolone, prednisone, beclomethasone dipropionate, triamcinolone, and mixtures thereof.
The anti-inflammatory cytokine that can be used as an additional treatment includes, but is not limited to, interleukin (IL)-1 receptor antagonist, IL-4, IL-6, IL-10, IL-11, and IL-13. Specific cytokine receptors for IL-1, tumor necrosis factor-alpha, and IL-18 also function as pro-inflammatory cytokine inhibitors. The nature of anti-inflammatory cytokines and soluble cytokine receptors are known in the art and discussed in Opal and DePalo, Chest, 117(4): 1162-72 (2000).
In some embodiments, the additional therapy includes one or more of: sulfasalazine, doxycycline, minocycline, penicillamine, tofacitinib, and leflunomide.
The components of the combination therapy may be administered substantially at the same time or sequentially.
The disclosure features a variety of methods for treating a viral disease or condition, (e.g., COVID-19) using the agents described herein.
As used herein, the term “treat” “treatment,” or “treating” a subject having an viral condition, are used in connection with a given treatment for a given disorder, wherein at least one symptom of the disorder is alleviated, or ameliorated. The treatment may inhibit deterioration or worsening of a symptom of the disclosed conditions (e.g., COVID-19) or may cause the condition to develop more slowly and/or to a lesser degree (e.g., fewer symptoms in the subject) in the subject than it would have absent the treatment. A subject is treated with the methods of this disclosure, to improve a condition, symptom, or parameter associated with a disorder or to prevent progression or exacerbation of the disorder (including secondary damage caused by the disorder) to either a statistically significant degree or to a degree detectable to one skilled in the art. A subject who is at risk for, diagnosed with, or who has one of the viral diseases and/or cytokine storn of this disclosure can be administered a compound of this disclosure (e.g., an agent that activates the UPR and/or an agent that disrupts the TCA cycle) in an amount and for a time to provide an overall therapeutic effect. A compound of this disclosure can be administered alone (monotherapy) or in combination with other agents (combination therapy), which agents are described in the “Additional treatments” section.
As used herein, the term “therapeutically effective amount” of an agent is an effective amount that may be determined by the effect of the administered agents or the combined effects of the agents (if more than one agent is used). The “therapeutically effective amount” of the agent of this disclosure is an amount that results in a reduction in the severity of disease symptoms, the frequency and length of periods without disease symptoms. Preferably, it results in prevention of dysfunction or disability due to an increase in disease or distress. For example, in the case of COVID-19, a therapeutically effective amount can be, for example, one that prevents or treats the cytokine storm. It is preferable to prevent further deterioration of physical symptoms associated with COVID-19. A therapeutically effective amount is also preferred to prevent or delay the onset of the cytokine storm, as may be desired when early or preliminary signs of disease (COVID-19) are present. Similarly, delaying the chronic progression associated with COVID-19 is also desired. Any clinical or biochemical test that monitors the above can be used to determine whether a particular treatment is in a therapeutically effective amount to treat COVID-19. Those skilled in the art will be able to determine such amounts based on factors such as the size of the subject, the severity of the subject's symptoms, and the particular composition or route of administration chosen.
The disclosed methods can also treat or prevent a COVID-19 cytokine storm by reducing the level of IL-Ib and/or TNF-a in the (blood or plasma) of the subject, preferably reducing the level of one of more, two or more, three or more, four or more, five or more or all six or TNF-a, IL-1B, IP-10, IL-6, IL-8, MCP-1, MIP-1a and MIP-Ib in (the blood or plasma) of the subject. The SARS-CoV-2 to be treated may either be the original wild-type strain and/or one or more variants. Examples of variants of SARS-CoV-2 that can be treated include, but are not limited to, the variants D614G, B.1.351, B.1.1.7, PI, P2, B.1.617, B.1.427, B.1.429, B.1.525 and B.1.526.
The therapeutically effective amount of the agent may also vary according to factors such as the disease state, the age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, e.g., to ameliorate at least one parameter of the condition or to ameliorate at least one symptom of the condition. A therapeutically effective amount is also an amount where the therapeutically beneficial effect exceeds any toxic or detrimental effect of the composition. A therapeutically effective amount of an agent of this disclosure (i.e., an effective dosage) includes milligram, microgram, nanogram, or picogram amounts of the agent per kilogram of subject or sample weight (e.g., about 1 nanogram per kilogram to about 500 micrograms per kilogram, about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).
The amounts and times of administration for combination therapies can be those that provide, e.g., an additive or a synergistic therapeutic effect. Further, the administration of the compound of this disclosure (e.g., a UPR activator and/or a TCA cycle inhibitor) can be used as a primary, e.g., first line treatment, or as a secondary treatment, e.g., for subjects who have an inadequate response to a previously administered therapy (i.e., a therapy other than one with a compound of this disclosure). In some embodiments, the combination therapy includes the use of a compound of this disclosure with one or more of the following agents: a corticosteroid, remdesivir, Nirmatrelvir, Bebtelovimab, Molnupiravir, an IL-6 inhibitor, an IL-1 inhibitor, a kinase inhibitor, a complement inhibitor, ivermectin, hydroxychloroquine, favipiravir, interferon-beta and a nonsteroidal anti-inflammatory drug (NSAID).
As used herein, the term “control” refers to an age-matched subject that does not have or is not diagnosed with a viral condition. In some embodiments, a control refers to an age-matched and sex-matched subject that is not treated with the method of this disclosure, or is treated with a placebo. In some embodiments, a control refers to a population average for the amount or degree of a particular parameter in a normal healthy population.
Treatment outcomes on viral diseases can be measured using any of the routine assays and techniques known in the art, including but not limited to enzyme-linked immunosorbent assay (ELISA), multiplex cytokines assay (Aziz N. Immunopathol Dis Therap. 2015; 6(1-2):19-22), qualitative and quantitative polymerase chain reaction (PCR), and patient-reported outcome measures. Clinical outcomes can be measured using several clinical features such as those described in Touma, Zahi (Ed.) Outcome Measures and Metrics in Systemic Lupus Erythematosus; Pages 1-50.
The methods of the disclosure can reduce production of inflammatory cytokines or chemokines by immune cells (such as dendritic cells) in the human subject. In some embodiments, the methods of this disclosure reduce IFN-I production in the human subject in need thereof by at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99%, as compared to the corresponding reference levels in the human subject or in a control. The methods can also reduce the expression of interferon stimulated genes, including, but not limited to, Guanylate Binding Protein 1 (GBP1), Interferon Regulatory Factor 7 (IRF7), interferon stimulated gene 54 (ISG54), myxovirus resistance protein B (MxB), and 2′-5′-Oligoadenylate Synthetase 2 (OAS2). The methods can also enhance expression of phosphoglycerate dehydrogenase (PHGDH), phosphoserine Phosphatase (PSPH), and phosphoserine Aminotransferase 1 (PSAT1). In some cases, the methods of the disclosure can reduce CXCL4 expression in DCs.
The practice of the methods and compositions of the disclosure employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), cell culture, immunology, cell biology, and biochemistry, which are well within the purview of the skilled artisan. Such techniques are explained in the literature, such as, “Molecular Cloning: A Laboratory Manual”, second edition (Sambrook, 1989); “Oligonucleotide Synthesis” (Gait, 1984); “Animal Cell Culture” (Freshney, 1987); “Methods in Enzymology” “Handbook of Experimental Immunology” (Weir, 1996); “Gene Transfer Vectors for Mammalian Cells” (Miller and Calos, 1987); “Current Protocols in Molecular Biology” (Ausubel, 1987); “PCR: The Polymerase Chain Reaction”, (Mullis, 1994); “Current Protocols in Immunology” (Coligan, 1991). These techniques are applicable to the methods and compositions of the disclosure. Particularly useful techniques for particular embodiments will be discussed in the sections that follow. The materials, reagents, and methods, further described below, are used in the following examples. The invention, as described in the following examples, do not limit the scope of the invention described in the claims.
Enriched leukocytes were obtained from the New York Blood Center (Long Island City, NY) after informed consent of donors who were deemed healthy by the New York Blood Center's criteria and used under a protocol approved by the Institutional Review Board of the Hospital for Special Surgery and the Institutional Biosafety Committee of Weill Cornell Medicine. The clinical and demographic characteristics of the donors reflect the diverse population of New York City. PBMCs were prepared using Ficoll-Paque density gradient (GE Healthcare) as previously described (32). pDCs and monocytes were isolated from PBMCs by positive selection using BDCA4-conjugated (33) and CD14-conjugated microbeads (Miltenyi Biotec), respectively. pDC depleted PBMC were prepared by removing BDCA4-positve cells from PBMC using microbeads (Miltenyi Biotec). Macrophages were differentiated from monocytes by culturing in complete RPMI 1640 medium with 20 ng/ml M-CSF for 1-5 d.
Human alveolar epithelial cells were obtained from ScienceCell and cultured in Alveolar Epithelial Cell Medium (ScienceCell) and expanded in T-25 plates and plated in Transwell plates (Corning) for additional experiments. Human alveolar macrophages were obtained from Accegen and were cultured in complete RPMI 1640 medium with 100 U/mL M-CSF, 1 mM Sodium Pyruvate, 10 mM HEPES, and 1× penicillin/streptomycin.
hPSC Culture The human induced pluripotent stem cells (iPSCs) were grown and maintained on 1% Matrigel-coated six-well plates in mTeSR (Stem Cell Technology) with 5% CO2 culture condition. The medium was changed daily. When iPSCs reached ˜90% confluence, the cells were passaged at 1:6-1:12 with Accutase (Stem Cell Technology).
hPSC Differentiation
Human induced pluripotent stem cells were differentiated into monocytes and macrophages as previously reported56. In brief iPSC were treated with accutase and scraped to produce small cellular groups and replated onto 1% Matrigel-coated 6-well plates. After 1-2 days, mTeSr medium was replaced with Mesoderm induction medium containing 15 ng/ml Activin A (R&D Systems), 40 ng/ml BMP-4 (R&D Systems), and 1.5 μm CHIR99021 (Stem Cell Technologies). On Day 2 the medium was replaced with hemogenic endothelium induction containing 50 ng/ml IL-6 (R&D Systems), 15 ng/ml IL-3 (R&D Systems), 50 ng/ml TPO (R&D Systems), 12.5 ng/ml bFGF (Peprotech), 50 ng/ml SCF (R&D Systems), and 50 ng/ml VEGF (R&D Systems). On Day 5 the medium was replaced with hematopoietic induction medium containing 50 ng/ml VEGF (R&D Systems), 50 ng/ml bFGF, 50 ng/ml SCF (R&D Systems) and 10 μM SB431542 (Tocris). On day 9 the cells were dissociated with Accutase and resuspended with Monocyte induction medium containing 50 ng/ml IL-6 (R&D Systems), 12 ng/ml IL-3 (R&D Systems), and 80 ng/ml M-CSF (R&D Systems) into low attachment plates. On day 15, floating cells were collected and StemSep Human CD14 Positive Selection Kit (Stem Cell Technologies) was used to isolated CD14+ cells. Macrophage were obtained by plating monocytes onto FBS-coated plates with macrophage differentiation media containing M-CSF. Cells were cultured under normoxic conditions at 37° C. and 5% CO2.
Activation of Cells For functional assay, freshly isolated pDCs were resuspended at 3.104 cells/100 ml complete RPMI and cultured in 96 wells/U bottom plate. PBMC and pDC-depleted PBMC were resuspended at 3×105 cells/100 ml complete RPMI and cultured in 96 wells/flat bottom plate. Untreated and gamma-irradiated inactivated SARS-CoV-2 (USA-WA1/2020, BEI resources) were used at a multiplicity of infection (MOI) of 1, 0.25, 0.5, 0.1 and 0.01 whereas Flu virus was used at MOI 2 for 3, 6, 10 and 18 hours. For blocking experiment, cells were pre-incubated for 1 h with the ACE2 inhibitor (Novus biologicals, 2 μM), TLR7 inhibitor IRS661 (2 μM), PI3Kδ inhibitor CAL-101 (10 μM), chlathrin inhibitor chlorpromazine (CPZ, 30 μM) or dynamin inhibitor dynasore hydrate (DH, 100 μM) followed by the addition of inactivated SARS-CoV-2 at MOI 0.25. After differentiation of macrophages, the cells were cultured at 1.105 cells/100 ml complete RPMI for 24 h either with 10% of non-activated pDC supernatant (Unst-pDC SN), 10% of SARS-CoV-2 activated pDC supernatant (SARS-pDC SN), recombinant IFN-α at 100000, 30000, 10000, 3000, 1000 and 300 μg/ml (PBL assay science) or inactivated SARS-COV-2 (MOI 0.25). LPS (2 or 10 ng/ml), Pam3Cys (20 ng/ml), PolyIC (10 ng/ml), ORN8L (60 μg/ml) or inactivated SARS-COV-2 (MOI0.25) were added in the culture for 6 h. For blocking experiments, macrophages were preincubated with JAK1/2 inhibitor baricitinib (2 μM) or anti-IFNAR antibody (2 μg/ml) for 1 h before adding of SARS-pDC SN for 24 h. TLR ligands were then added to the well for 6 h. For TNF blocking, SARS-pDC SN and macrophages were pre-incubated for 1 hour with anti-hTNF (10 μg/ml, R&D Systems) and anti-hTNFRI (10 μg/ml, R&D Systems) respectively. After 1 hour, macrophages were incubated with SARS-pDC SN+ anti-hTNF for 24 hours. LPS (10 ng/ml) was then added to the well for 6 h.
2×105 human alveolar epithelial cells were plated in regular Tissue Culture plates in Alveolar Epithelial Cell Medium supplemented with 100 U/mL M-CSF while 5.104 human alveolar macrophages were plated in Transwell Plate Inserts in Alveolar Epithelial Cell Medium supplemented with 100 U/mL M-CSF. Cultures were inoculated with MOCK infection or SARSCoV-2 (USA-WA1/2020, BEI resources) at MOI=0.1. After infections cells were lysed in Trizol or fixed in 4% paraformaldehyde for 24 hrs. For co-culture cells, cells were lifted with trypsin and then fixed in 4% paraformaldehyde for 24 hrs. Macrophages were then isolated and separated used anti-human CD68 antibody (Stem Cell Technology) and StemSep beads (Stem Cell Technology).
SARS-CoV-2 isolate USA-WA1/2020 (NR-52281) was provided by the Center for Disease Control and Prevention and obtained through BEI Resources, NIAID, NIH. SARS-CoV-2 was propagated in Vero E6 cells in DMEM supplemented with 2% FBS, 4.5 g/L D-glucose, 4 mM Lglutamine, 10 mM Non-essential amino acids, 1 mM sodium pyruvate and 10 mM HEPES using a passage-2 stock of virus as described previously57. Three days after infection virus-containing supernatants were purified as described previously (Nilsson-Payant, B. E. et al. J Virol 95, e0125721). In brief, supernatant containing propagated virus was filtered through an Amicon Ultra 15 (100 kDa) centrifugal filter (Millipore Sigma) at ˜4000 rpm for 20 minutes. Flow through was discarded and virus was resuspended in DMEM supplemented as described above. Infectious titers of SARS-CoV-2 were determined by plaque assay in Vero E6 cells in Minimum Essential Media supplemented with 2% FBS, 4 mM Lglutamine, 0.2% BSA, 10 mM HEPES and 0.12% NaHCO3 and 0.7% agar. All MOIs were based on titer determined from plaque assays on Vero E6 cells. All work involving live SARS-CoV-2 was performed in the CDC and USDA-approved BSL-3 facility of the Icahn School of Medicine at Mount Sinai and NYU Langone in accordance with institutional biosafety requirements.
PCR reactions were performed as described previously with 10 ng of cDNA. In brief, RNA was extracted from cells using the Qiagen RNeasy Mini Kit. Quantity of RNA was measured by Nanodrop, and high-capacity cDNA Reverse Transcription kit was used to generate 20-50 ng cDNA. Gene expression levels were calculated based on relative threshold cycle (Ct) values. This was done using the formula Relative Ct=100×1.8 (HSK-GENE), where HSK is the mean CT of duplicate housekeeping gene runs (Ubiquitin), GENE is the mean CT of duplicate runs of the gene of interest, and 100 is arbitrarily chosen as a factor to bring all values above 0. Primers were from Fischer Scientific.
Total RNA samples were prepared from cells using TRIzol and the Direct-zol RNA Miniprep Plus kit (Zymo Research) according to the manufacturer's instructions. To quantify viral replication, measured by the accumulation of subgenomic N transcripts, one-step quantitative real-time PCR was performed using SuperScript III Platinum SYBR Green One-Step qRT-PCR Kit (Invitrogen) with primers specific for the TRS-L and TRS-B sites for the N gene as well as 18S and ACTB as an internal reference as previously described59. Quantitative real-time PCR reactions were performed on the Biorad CFX384 Touch Real-Time PCR Detection System. Delta-delta-cycle threshold (DDCT) was determined relative to the 18S and ACTB and mock infected/treated samples.
Supernatant from pDCs 24 h culture was used for quantification of secreted TNF and IL-6 and measured with the use of an enzyme-linked immunosorbent assay (ELISA) according to the manufacturer's protocol (Mabtech).
Cells were lysed in 50 μl of cold 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% (vol/vol) Triton X-100, 2 mM Na3VO4, 1× phosSTOP EASYPACK, 1 mM Pefabloc, and 1×EDTA-free complete protease inhibitor cocktail (Roche, Basel, Switzerland), and incubated for 10 min on ice. Then, cell debris was pelleted at 13,000 rpm at 4-C for 10 min. The soluble protein fraction was mixed with 4× Laemmli Sample buffer (BIO-RAD, Cat. #1610747) and 2-mercroptoehanol (BME) (Sigma-Aldrich). Samples for Western Blot were subjected to electrophoresis on 4-12% Bis-Tris gels (Invitrogen). To detect IRF5 dimers, Novex WedgeWell 14% Tris-Glycine Gel (Invitrogen, Cat. #XP00140BOX) was adopted to electrophoresis of protein samples according to manufacturer's instruction. Proteins were transferred to polyvinylidene difluoride membrane and immunodetection was performed as previously published 87. Antibodies used are from Cell Signaling Technology: IkB-α (9242s), phosphor-p38 (9215S), p38 (9212S) and Phospho-p44/42 μMAP Kinase (Erk1/2) (9101S).
To evaluate inflammatory gene in the genome associated with regulatory activity, FAIER assay was adopted as previous study60. Briefly, cells were cross-lined with 1% formaldehyde for 15 min and quenched with 0.125 μM glycine. Then, cells were lysed and sonicated. 10% of the samples were used for input and the rest for phenol/chloroform extraction. The input DNA and extracted DNA were used for qPCR.
After RNA extraction, libraries for sequencing were prepared using the NEBNext® Ultra™ II RNA Library Prep Kit for Illumina® following the manufacturer's instructions (Illumina). Quality of all RNA and library preparations were evaluated with BioAnalyser 2100 (Agilent) and the sequencing input was 500 ng total RNA. Sequencing libraries were sequenced by the Genomics Facility at Weill Cornell using a NextSeq2000, 50-bp paired-end reads at an average depth of 35 million reads per sample.
Read quality was assessed and adapters trimmed using fastp. Reads were then mapped to the human genome (hg38) and reads in exons were counted against Gencode version 33 (Ensembl 99) with STAR Aligner. Differential gene expression analysis was performed in R (4) using edgeR. Genes with low expression levels (<4 counts per million in at least one group) were filtered from all downstream analyses. Benjamini-Hochberg false discovery rate (FDR) procedure was used to correct for multiple testing. Downstream analyses were performed in R using a visualization platform built with Shiny developed by bioinformaticians at the David Z. Rosensweig Genomics Research Center at the Hospital for Special Surgery.
Analysis of Single-Nucleus RNA Sequencing Data from Lungs of COVID-19 Patients
Publicly available data of single-nucleus RNA sequencing (snRNA-seq) was used from postmortem lung tissue of COVID-19 patients4. Both the original global UMAP representation and cell type labels provided by authors were used. Since pDCs were not labeled as a cell type, dendritic cells were re-clustered by re-calculating a neighbor graph and computing a UMAP representation with only those cells using default parameters of Scanpy (version 1.8.1), as described (Melms, J. C. et al. Nature 595, 114-119, (2021). Dendritic cells were then clustered using the Leiden algorithm with resolution 0.4. The top representative genes for each cluster were obtained using the “rank_genes_groups” function of Scanpy on the original raw expression values and with a T-test overestimating the variance of each group. Gene expression displayed as overlay to UMAP plots or in heatmaps represents raw counts log transformed after adding the unit value. To test the under- or over-representation of cells of either Control or COVID-19 patients within each cluster, Fisher's exact test was used with a two-sided alternative and multiple testing correction using Benjamini-Hochberg's false discovery rate method in Pingouin (version 0.4.0), as described4.
To investigate the cellular state of each cell type dependent on COVID-19 infection, two sources of gene expression signatures were used: i) genes up-regulated in association with inflammation and fibrotic phenotype in COVID-19 discovered using bulk RNA sequencing; ii) the set of 50 Hallmark pathways from the Molecular Signatures database (MsigDB, version 7.4), as described4.
Each signature was scored in every single cell using the “score_genes” function of Scanpy, and the values were aggregate per cell type and disease state (Control/COVID-19) using the mean. The difference between the values in COVID-19 compared with Control in each cell type was used as a measure of differential signature activity associated with disease. Furthermore, to estimate the relationship between interferon alpha, inflammation and fibrosis during COVID-19, the Pearson correlation was calculated between these signatures across cells of the same cell type and disease state, for all cell types. This was done with equal cell numbers per cell type in Control and COVID-19 by randomly sampling the same number of cells.
Graphpad Prism for Windows was applied for all statistical analysis. The data are shown as means±SME. Detailed information about statistical analysis, including tests and values used, and number of times experiments is provided in the figure legends.
Lung samples were analyzed from patients with terminal COVID-19 (Melms, J. C. et al. Nature 595, 114-119, (2021); Rendeiro, A. F. et al. Nature 593, 564-569, (2021) and cells from bronchoalveolar lavage (BAL) of patients with mild or severe forms of COVID-19 were also analyzed. In this latter population, a large population of macrophages (
The relationship between IFN-I and inflammatory responses was analyzed in macrophages at the single-cell level and the two programs were found to be anti-correlated in controls, but correlated in COVID-19 patients, with macrophages from severe patients showing the highest correlation (
There was found to be a strong correlation between the IFN-I response and the presence of pro-fibrotic signals in macrophages. This observation led to the hypothesis that the heterogeneous information contained in the scRNA-seq data can be used to infer a pseudo-temporal continuum of the molecular response to SARS-CoV-2 infection in macrophages. Without being bound by theory, it is believed that patients at different stages of the disease could give an indication of the progression of the disease in SARS-CoV-2-infected patients. Using diffusion maps on the transcriptome data of macrophages, a joint representation was inferred which first dimension is associated with the dynamics of disease progression (
This unsupervised, dynamic view of macrophage response reinforces the idea of sequential stages of activation of the macrophages during COVID, associated with the early induction of IFN-I which primes macrophages for hyperinflammatory activation in a subset of patients which develop severe disease. To determine the source of the IFN-I, the dataset was analyzed and the infiltration of the BAL by pDCs in patients with mild disease was observed (
These pDCs were found to express IFN-I-regulated genes (
The cellular composition and activation status of lung cells in recently deceased patients following SARS-CoV-2 infection has been described (Melms, J. C. et al. Nature 595, 114-119, (2021); Rendeiro, A. F. et al. Nature 593, 564-569 (2021)). By conducting subcluster analysis in the dendritic cell subpopulation, pDCs were identified (
A similar positive correlation was also observed at the pathway level between the IFN pathway and both the inflammation and fibrotic pathways (
These data would indicate that during the course of the disease, the intense priming of macrophages by pDC-derived IFN-I, which likely occurs early in the disease pathogenesis, can prime macrophages and in particular monocyte-derived macrophages.
Although IFN-180 I is critical for the clinical response to SARS-CoV-2 infection, the cellular source of IFN-I is not well defined. Purified pDCs were found to have a robust IFN-α response to both live, but also UV-inactivated SARS-CoV-2 (
It has been described that less than 10% of lung macrophages are infected with SARS-CoV-2 (Rendeiro, A. F. et al. Nature 593, 564-569, (2021). However, it was recently reported that macrophages produce some IFN-I due to the activation of the cGAS-STING pathway (Lore, K., et al. J. Exp Med. 201, 2023-2033 (2005)), as the deletion of STING in a mouse model of SARS infection partially reduced IFN-I and ISG expression. However, the authors did not observe that macrophages can be directly stimulated by SARS-CoV-2.
Hence, it was observed that neither CD14+ monocytes, pluripotent stem cells (PSCs) derived macrophages, monocyte derived macrophages or primary alveolar macrophages isolated from human lungs can directly be infected nor stimulated by live SARS-CoV-2 (
Combined with the in vitro data of live SARS-CoV-2 inoculated pDCs, these data support a scenario where the IFN-I is coming from at least 2 different sources—pDCs by directly sensing of the live virus and macrophages by interacting with epithelial cells infected by the virus (see model
An increase in bacterial infections in patients with COVID-19, resulting in higher levels of bacterial products (bacterial DNA/RNA, lipoproteins and also of LPS) in ICU patients has been reported (Arunachalam, P. S. et al. Science 369, 1210-1220, (2020)). Thus, it was determined whether SARS-CoV-2 activated pDCs, especially IFN-I induced in these cells, could impact macrophage response into the secretion of the so-called cytokine storm (Rendeiro, A. F. et al. Nature 593, 564-569, (2021)). Supernatants of SARS-pDCs had little impact when used alone but drastically amplified the production and expression of proinflammatory cytokines such as TNF and TL-6 by macrophages in response to LPS but also to Pam3Cys (agonist of another transmembrane TLR), Poly I.C, and the TLR8 agonist ORN8L (both being RNA-sensing TLRs which are endosomal) (
As pDCs produce large amounts of IFN-I in response to SARS-CoV-2 (
To obtain a comprehensive understanding of how type I IFN and SARS-pDC supernatants influence macrophage activation, transcriptomic analysis was performed using RNA-seq to evaluate TLR4 responses in macrophages exposed with IFN-α or SARS-pDC supernatants. Principal component analysis (PCA) showed that pDC supernatants and IFN-α conditions closely clustered together (
Accordingly, inhibition of IFN-I production using either the TLR7 inhibitor IRS661 or the PI3Kδ inhibitor CAL-101 reduced inflammatory gene induction upon challenge with LPS (
The spliced XBP1 isoform generated by IRE1a encodes the functional transcription factor XBP1, which induces factors implicated in restoring ER proteostasis while controlling diverse metabolic programs (25, 26).
For the following experiments, enriched leukocytes were obtained from New York blood center (Long Island City, NY) under internal Institutional Review Board-approved protocols. PBMCs were prepared using Ficoll-Paque density gradient and pDCs were isolated using BDCA4+ positive selection (Miltenyi Biotech: 130-097-415) as previously described (42). pDCs were cultured at 40,000 cells (for healthy donors) per well in a 96-round bottom plate and incubated at 37° C., 5% CO2 and 95% humidity. For TLR7 and TLR9 activation assay, pDCs were stimulated with heat-inactivated 2 μMOI of H1N1 VR-95 influenza A virus (ATCC) and 0.075 μM of C274 (42) respectively.
In some culture conditions, cells were cultured with the tunicamycin (thermofisher: 654380), thapsigargin (Sigma: T9033), 4 μ8c (EMD Millipore: 412512), MKC8866 (Medchem Express: HY-104040), IXA4 (Chembridge: 131171.1), AMG PERK44 (R&D: 5517), Ceapin-A7 (Sigma: SML2330), NCT-503(Axon Medchem: 2623), L-serine (EMD Millipore: S4500), sodium pyruvate (Sigma: 8636), α-ketoglutaric acid disodium salt hydrate (Sigma: K3752), CPI-613 (Selleckchem: S2776), Anti-PF4 antibody (Abcam: ab9561), CXCL4 (Sigma: SRP3142).
Using gene set enrichment analysis (GSEA), it was observed that transcriptional networks implicated in amino acid biosynthesis were markedly activated in pDCs experiencing ER stress, with or without TLR9 agonist treatment (
Using an inhibitor of both α-ketoglutarate dehydrogenase (KGDH) and pyruvate dehydrogenase (PDH), called CPI-613 (6,8-bis-benzylthio-octanoic acid), it was tested whether disrupting the TCA cycle could impact the IFN-α response by pDCs. CPI-613 has been well characterized and is in clinical trials for pancreatic cancer (36, 37). As shown in
Collectively, these data indicate that pyruvate and α-KG are key intermediate metabolites in the TCA cycle that are required for optimal IFN-α responses in TLR9-activated pDCs, and that this process is markedly blunted upon ER stress-driven activation of RE1-XBP1 signaling due to the increased activity of PHGDH.
It is expected that a TCA inhibitor will reduce IFNA expression in the lungs of COVID-19 subjects and in pDCs isolated from the lungs of COVID-19 subjects. Inhibitors of the TCA cycle are shown in
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
This application claims the benefit of the filing date of U.S. Provisional Application No. 63/255,336, filed Oct. 13, 2021, and U.S. Provisional Application No. 63/347,715, filed Jun. 1, 2022, the contents of which are incorporated by reference in their entirety herein.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2022/046404 | 10/12/2022 | WO |
Number | Date | Country | |
---|---|---|---|
63347715 | Jun 2022 | US | |
63255336 | Oct 2021 | US |