Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INTERFERON-BETA ANALOG PEPTIDE
Document Type and Number:
WIPO Patent Application WO/2019/073315
Kind Code:
A1
Abstract:
Disclosed herein is an interferon-beta (IFNβ) analog peptide including a plurality of amino acid substitutions in an amino acid sequence of IFNβ as set forth in SEQ ID No. 4. The IFNβ analog peptide may include a plurality of amino acid substitution in the amino acid sequence of IFNβ in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combinations thereof, numbered in accordance with SEQ ID No. 4.

Inventors:
POOREBRAHIM MANSOUR (IR)
ASGHARI MATIN (IR)
NASR-ESFAHANI MOHAMMAD HOSSEIN (IR)
SANADGOL NIMA (IR)
ABAZARI MOHAMMAD FOAD (IR)
NOURI ALEAGHA MARYAM (IR)
ASKARI HASSAN (IR)
SADEGHI SOLMAZ (IR)
Application Number:
PCT/IB2018/056577
Publication Date:
April 18, 2019
Filing Date:
August 29, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
POOREBRAHIM MANSOUR (IR)
International Classes:
C07K14/565
Domestic Patent References:
WO2007110231A22007-10-04
Attorney, Agent or Firm:
IDESAZAN ASR AFTAB (IR)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. An interferon-beta (IFNP) analog peptide comprising a plurality of amino acid substitutions in an amino acid sequence of IFNP as set forth in SEQ ID No. 4, the IFNP analog peptide comprising a plurality of amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combinations thereof, numbered in accordance with SEQ ID No. 4.

2. The IFNP analog peptide according to claim 1 , wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5F, F6R, I8G, F9W, R10H, D12N, S13T, S14L, S15N, W18L, N19R, E20W, T21V, I22G, V23F, and combinations thereof.

3. The IFNP analog peptide according to claim 2, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 1.

4. The IFNP analog peptide according to claim 1 , wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5G, F6R, A7D, I8V, F9R, R10K, Q11R, D12G, S13F, S 15D, W18L, N19R, E20K, T21L, I22G, V23R, E24V, and combinations thereof

5. The IFNP analog peptide according to claim 4, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 2.

6. The IFNP analog peptide according to claim 1 , wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5G, F6R, I8L, F9R, R10K, D12N, S13F, S14L, S15N, W18L, N19R, E20K, T21L, I22G, V23H, and combinations thereof.

7. The IFNP analog peptide according to claim 6, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 3.

8. The IFNP analog peptide according to claim 1, wherein the IFNP analog peptide comprises 27 amino acids.

9. The IFNP analog peptide according to claim 1, wherein the IFNP analog peptide has an antiviral activity and an immunomodulatory activity,

wherein the immunomodulatory activity comprises suppressing pro-inflammation mediators.

10. A method for treating multiple sclerosis (MS) or viral infections in a patient, comprising: administering an effective amount of an IFNP analog peptide, the IFNP analog peptide comprising a plurality of amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24 and combinations thereof, of an amino acid sequence of IFNP, numbered in accordance with SEQ ID No. 4.

11. The method according to claim 10, wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5F, F6R, I8G, F9W, R10H, D12N, S13T, S14L, S15N, W18L, N19R, E20W, T21V, I22G, V23F, and combinations thereof.

12. The method according to claim 10, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 1.

13. The method according to claim 10, wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5G, F6R, A7D, I8V, F9R, R10K, Q11R, D12G, S13F, S 15D, W18L, N19R, E20K, T21L, I22G, V23R, E24V, and combinations thereof

14. The method according to claim 10, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 2.

15. The method according to claim 10, wherein the IFNP analog peptide comprises a plurality of amino acid substitutions in the amino acid sequence of IFNP, the amino acid substitutions comprise at least one of I5G, F6R, I8L, F9R, R10K, D12N, S13F, S14L, S15N, W18L, N19R, E20K, T21L, I22G, V23H, and combinations thereof.

16. The method according to claim 10, wherein the IFNP analog peptide has an amino acid sequence as set forth in SEQ ID No. 3.

17. The method according to claim 10, wherein the IFNP analog peptide comprises 27 amino acids.

18. The method according to claim 10, wherein the effective amount of the IFNP analog peptide is between 10 mg/kg of body weight and 20 mg kg of body weight.

19. The method according to claim 10, administering the effective amount of an IFNP analog peptide comprises intravenous injection of the effective amount of an IFNP analog peptide to the patient.

Description:
INTERFERON-BETA ANALOG PEPTIDE

STATEMENT REGARDING SEQUENCE LISTING

[0001] The Sequence Listing associated with this application is provided in text format in lieu of a paper copy and is hereby incorporated by reference into the specification. The name of the text file containing the Sequence Listing is (050-3266) Sequence Listing.txt. The text file is 2 KB, was created on August 29, 2018, and is being submitted electronically via ePCT.

TECHNICAL FIELD

[0002] The present disclosure generally relates to peptide-based drugs, particularly to interferon beta (IFNP) analog peptides, and more particularly to IFNP analog peptides for treatment of multiple sclerosis (MS).

BACKGROUND ART

[0003] Multiple sclerosis (MS) is an autoimmune disease which may attack the myelin proteins in the central nervous system (CNS). One of the most important hallmarks of the MS process is that peripherally activated inflammatory cells may pass across the blood-brain barrier (BBB) leading to demyelination within the CNS. Numerous studies have been undertaken to understand the key inflammatory mediators and neurodegenerative mechanisms that underlie the relapsing and progressive phase of the disease.

[0004] Interferon-beta (IFNP)-based formulations have been shown to alleviate the exacerbations of MS and may be used as a first-line treatment for relap sing-remitting multiple sclerosis (RRMS) (WO/2002/036628A2, US20090311216 Al). However, IFNp-based therapy may lead to a number of adverse side effects including flu-like symptoms, inconsistencies in patient laboratory analyses, menstrual disorders, and increased spasticity. Moreover, similar to other clinically available protein-based drugs, IFNP has several drawbacks including a high number of antigenic regions, high cost, and susceptibility to proteolytic degeneration.

[0005] Peptide-based drugs have several unique physicochemical properties that make them attractive in medical interventions. Since peptides may be suitable alternatives of IFNP for treatment of MS, there is a need for an IFNP analog peptide with improved physicochemical properties, lower number of antigenic regions, lower cost, and higher stability. Moreover, there is a need for an IFNP analog peptide for the treatment of MS without IFNP side effects. SUMMARY OF THE DISCLOSURE

[0006] This summary is intended to provide an overview of the subject matter of this patent, and is not intended to identify essential elements or key elements of the subject matter, nor is it intended to be used to determine the scope of the claimed implementations. The proper scope of this patent may be ascertained from the claims set forth below in view of the detailed description below and the drawings.

[0007] In one general aspect, the present disclosure describes an exemplary interferon-beta (IFNP) analog peptide including a plurality of amino acid substitutions in an amino acid sequence of IFNP as set forth in SEQ ID No. 4. The IFNP analog peptide may include a plurality of amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combinations thereof, numbered in accordance with SEQ ID No. 4.

[0008] The above general aspect may include one or more of the following features. In some exemplary embodiments, the IFNP analog peptide may include at least one of I5F, F6R, I8G, F9W, R10H, D12N, S 13T, S 14L, S 15N, W18L, N19R, E20W, T21V, I22G, V23F amino acid substitutions, and combinations thereof, in the amino acid sequence of IFNp. In some exemplary embodiments, the IFNP analog peptide may have an amino acid sequence as set forth in SEQ ID No. 1.

[0009] According to some exemplary embodiments, the IFNP analog peptide may include at least one of I5G, F6R, A7D, I8V, F9R, R10K, Ql 1R, D12G, S 13F, S 15D, W18L, N19R, E20K, T21L, I22G, V23R, E24V amino acid substitutions, and combinations thereof in the amino acid sequence of IFNP with SEQ ID No. 4. In some exemplary embodiments, the IFNP analog peptide may have an amino acid sequence as set forth in SEQ ID No. 2

[0010] According to some exemplary embodiments, the IFNP analog peptide may include at least one of I5G, F6R, I8L, F9R, R10K, D12N, S 13F, S 14L, S 15N, W18L, N19R, E20K, T21L, I22G, V23H amino acid substitutions, and combinations thereof in the amino acid sequence of IFNP with SEQ ID No. 4. In some exemplary embodiments, the IFNP analog peptide may have an amino acid sequence as set forth in SEQ ID No. 3. In some exemplary embodiments, the IFNP analog peptide may include about 27 amino acids. In some exemplary embodiments, the IFNP analog peptide may have an antiviral activity and an immunomodulatory activity including suppressing pro-inflammation mediators. [0011] In another general aspect, the present disclosure describes an exemplary method for treating multiple sclerosis (MS) and viral infections in a patient. The method may include administering an effective amount of the IFNP analog peptide. The IFNP analog peptide may include a plurality of amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combination thereof, numbered in accordance with SEQ ID No. 4.

[0012] According to some exemplary embodiments, the effective amount of the IFNP analog peptide may be between about 10 mg/kg of body weight and about 20 mg/kg of body weight. In some exemplary embodiments, administering the effective amount of the IFNP analog peptide may include intravenously injecting the effective amount of the IFNP analog peptide to the patient.

BRIEF DESCRIPTION OF THE DRAWINGS

[0013] The drawing figures depict one or more implementations in accord with the present teachings, by way of example only, not by way of limitation. In the figures, like reference numerals refer to the same or similar elements.

[0014] FIG. 1 illustrates a method for producing an interferon-beta (IFNP) analog peptide, consistent with one or more exemplary of the present disclosure.

[0015] FIG. 2A illustrates a representation of IFNP structure with cross-species conservation scores, consistent with one or more exemplary embodiments of the present disclosure.

[0016] FIG. 2B illustrates a representation of a highly conserved region of IFNP, consistent with one or more exemplary embodiments of the present disclosure.

[0017] FIG. 2C illustrates a human IFNP-interferon-alpha/beta receptor alpha chain (IFNAR1) complex after molecular docking, consistent with one or more exemplary embodiments of the present disclosure

[0018] FIG. 3A illustrates an interaction mode between a highly conserved peptide and IFNAR1 after a high-resolution peptide -protein docking, consistent with one or more exemplary embodiments of the present disclosure.

[0019] FIG. 3B illustrates an interaction mode between the IFNP analog peptide and IFNAR1 after a high-resolution peptide -protein docking, consistent with one or more exemplary embodiments of the present disclosure. [0020] FIG. 4A illustrates a plotted root mean square deviation (RMSD) plot for complexes of different peptides with IFNARl, consistent with one or more exemplary embodiments of the present disclosure.

[0021] FIG. 4B illustrates a radius of gyration (Rg) plot for complexes of different peptides with IFNARl, consistent with one or more exemplary embodiments of the present disclosure.

[0022] FIG. 5A illustrates fluorescence emission spectra of IFNARl at different concentrations of an ΠΤΝΓβ analog peptide, consistent with one or more exemplary embodiments of the present disclosure.

[0023] FIG. 5B illustrates a plot for quenching of IFNARl intrinsic fluorescent by an IFNP analog peptide, consistent with one or more exemplary embodiments of the present disclosure.

[0024] FIG. 6 illustrates mean clinical scores of different experimental groups after immunization, consistent with one or more exemplary of the present disclosure.

[0025] FIG. 7A illustrates an expression fold change of matrix metalloproteinase 2 (MMP2) in ΠΤΝΓβ analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure.

[0026] FIG. 7B illustrates an expression fold change of matrix metalloproteinase 9 (MMP9) and MMP2 in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure.

[0027] FIG. 7C illustrates a concentration change of tumor necrosis factor alpha (T Fa) in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure.

[0028] FIG. 7D illustrates a concentration change of interleukin 1 beta (ILip) in ΠΤΝΓβ analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure.

[0029] FIG. 8A illustrates a percentage of interleukin 17 (IL17) positive cells, cluster of differentiation l ib (CD l ib) positive cells, and cluster of differentiation 45 (CD45) positive cells in each μιη2 of mice cerebral cortex after injection of an ΠΤΝΓβ analog peptide, consistent with one or more exemplary embodiments of the present disclosure.

[0030] FIG. 8B illustrates a percentage of IL17, CD l ib and CD45 positive cells in each μιη 2 of mice cerebral cortex after IFNP injection, consistent with one or more exemplary embodiments of the present disclosure. DESCRIPTION OF EMBODIMENTS

[0031] In the following detailed description, numerous specific details are set forth by way of examples in order to provide a thorough understanding of the relevant teachings. However, it should be apparent that the present teachings may be practiced without such details. In other instances, well known methods, procedures, components, and/or circuitry have been described at a relatively high-level, without detail, in order to avoid unnecessarily obscuring aspects of the present teachings.

[0032] Interferons may initiate specific signals via formation of heterodimeric IFNAR1- IFNAR2 complexes. However, interferon-beta (ΠΤΝΓβ) may be able to induce intracellular signals through binding to IFNAR1, independent of interferon- alpha/beta receptor beta chain (IFNAR2). Thus, IFNP may be a promising candidate for designing analog peptides in MS therapy without targeting the IFNAR2. However, efficiency of IFNP-based drugs may be considerably limited due to their undesirable properties, especially high immunogenicity.

[0033] In the present disclosure, an exemplary ΠΤΝΓβ analog peptide may be capable of significantly reducing brain dysfunction in MS by down-regulating production of inflammatory mediators. Disclosed herein is the exemplary ΠΤΝΓβ analog peptide including a plurality of amino acid substitutions in an amino acid sequence of ΠΤΝΓβ as set forth in SEQ ID No. 4. The IFNP analog peptide may include a plurality of amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combinations thereof, numbered in accordance with SEQ ID No. 4.

[0034] FIG. 1 shows method 100 for identifying the exemplary IFNP analog peptide, consistent with one or more exemplary embodiments of the present disclosure. Method 100 may include identifying a highly conserved peptide in an amino acid sequence of ΠΤΝΓβ (step 102), generating a library of tolerated peptides by substituting an amino acid in the highly conserved peptide (step 104), and selecting an exemplary ΠΤΝΓβ analog peptide by improving binding affinity and stability of the tolerated peptides (step 106).

[0035] Step 102 may include identifying the highly conserved peptide in the amino acid sequence of ΠΤΝΓβ. In some exemplary implementations, identifying the highly conserved peptide in the amino acid sequence of ΠΤΝΓβ may include identifying conserved residues in the amino acid sequence of IFN-β, predicting functional residues of IFN-β, determining mode of interaction of an ΙΡΝβ-IFNARl complex, and identifying functionally interacting residues of ΙΡΝβ and IFNAR1. [0036] In some exemplary implementations, the highly conserved peptide in the amino acid sequence of ΠΤΝΓβ may be identified using a multiple sequence alignment (MSA) of human IFNP, mouse IFNP, and rat IFNp. In some exemplary embodiments, the MSA may be conducted using at least one of Clustal-W program, ConSurf program, or combinations thereof. In some exemplary implementations, the functional residues of IFNP which may be involved in IFNP activity may be predicted by identifying highly conserved patches of three-dimensional (3D) structure of the IFNp. In some exemplary embodiments, the functional residues of IFNP may be predicted using the ConSurf program and molecular docking.

[0037] In some exemplary embodiments, the mode of interaction of the human IFNP-IFNAR1 complex may be determined by evaluating the mode of interaction for the murine IfnP-Ifnarl complex and identifying the structurally conserved residues between the human and murine proteins. In some exemplary embodiments, the human IFNP and murine IfnP may be structurally aligned using SuperPose and TM-align servers. In some exemplary embodiments, structures of human IFNARl and murine Ifnarl may be aligned using SuperPose and TM-align servers.

[0038] In some exemplary implementations, identifying the functionally interacting residues of IFNP and IFNARl may include determining a ligand-binding site of human IFNARl and an IFNAR-binding site of human IFNp. In some exemplary implementations, the functionally interacting residues of IFNP and IFNARl may be identified using molecular docking and choosing protein complexes with the lowest binding energy. In some exemplary embodiments, molecular docking may be done utilizing at least one of ClusPro web tool, HADDOCK web tool, FlexPepDock web tool, and combinations thereof. In some exemplary embodiments, the highly conserved peptide may include about 27 amino acids of the IFNP at positions from about 83 to about 109. In some exemplary embodiments, the highly conserved peptide may have an amino acid sequence as set forth in SEQ ID No. 4 and may be one of the most important regions involved in interaction of IFNP with IFNARl .

[0039] Step 104 may include generating the library of tolerated peptides by substituting the amino acid in the highly conserved sequence. In some exemplary embodiments, the library of tolerated peptides may be generated using Backrub and sequence tolerance protocols implemented in Rosetta package based on IFNARl -binding site of the IFNp. It should be noted that functional residues involved in interaction with IFNARl and in the biological activity of the ΠΤΝΓβ may not be substituted. In some exemplary embodiments, the most tolerated peptides may be selected from the library of tolerated peptides by in-silico screening.

[0040] In some exemplary implementations, substituting the amino acid in the highly conserved peptide may lead to improving physicochemical properties of the highly conserved sequence. In one or more exemplary embodiments, the physicochemical properties may include molecular weight, theoretical isoelectric point (pi), net charge at pH level about 7, instability index, grand average of hydropathicity (GRAVY), number of aggregation hot spots, number of antigenic regions, half -life, water solubility, and cell-penetrating capability.

[0041] Step 106 may include selecting the IFNP analog peptide by improving binding affinity and stability of the tolerated peptides. In some exemplary implementations, selecting the IFNP analog peptide may include selecting candidate peptides by improving binding affinity of the tolerated peptide and selecting the most stable candidate peptides by calculating free energy of the candidate peptides.

[0042] In some exemplary implementations, improving binding affinity of the tolerated peptides may include docking the tolerated peptide with IFNARl and selecting candidate peptides with high binding affinity. In some exemplary implementations, improving stability of the tolerated peptides may include calculating free energy and selecting the most stable candidate peptides.

[0043] In some exemplary embodiments, the ΠΤΝΓβ analog peptide may include the amino acid substitution in at least one position of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 19, 20, 21, 22, 23, 24, and combinations thereof, numbered in accordance with SEQ ID No. 4. In some exemplary embodiments, the IFNP analog peptide may include the amino acid substitution in the amino acid sequence of the IFNp including at least one of I5F, F6R, I8G, F9W, R10H, D12N, S13T, S 14L, S 15N, W18L, N19R, E20W, T21V, I22G, V23F, and combinations thereof. In some exemplary embodiments, the ΠΤΝΓβ analog peptide may have an amino acid sequence as set forth in SEQ ID No. 1.

[0044] In some exemplary embodiments, the ΠΤΝΓβ analog peptide may include the amino acid substitution in the amino acid sequence of the IFNP including at least one of I5G, F6R, A7D, I8V, F9R, R10K, Q11R, D12G, S 13F, S 15D, W18L, N19R, E20K, T21L, I22G, V23R, E24V, and combinations thereof. In some exemplary embodiments, the IFNP analog peptide may have an amino acid sequence as set forth in SEQ ID No. 2. [0045] In some exemplary embodiments, the ΠΤΝΓβ analog peptide may include the amino acid substitution in the amino acid sequence of the ΠΤΝΓβ including at least one of I5G, F6R, I8L, F9R, R10K, D12N, S 13F, S 14L, S 15N, W18L, N19R, E20K, T21L, I22G, V23H, and combinations thereof. In some exemplary embodiments, the ΠΤΝΓβ analog peptide may have an amino acid sequence as set forth in SEQ ID No. 3. In some exemplary embodiments, the IFNP analog peptide may include 27 amino acids.

[0046] In some exemplary embodiments, the ΠΤΝΓβ analog peptide may have an antiviral activity and an immunomodulatory activity including suppressing pro -inflammation mediators. In some exemplary embodiments, the ΠΤΝΓβ analog peptide may be used for treating multiple sclerosis (MS) and viral infections in a patient by administering an effective amount of an ΠΤΝΓβ analog peptide.

[0047] In some exemplary embodiments, the ΙΡΝβ may be used for treating relapsing-remitting multiple sclerosis (RRMS). In some exemplary embodiments, the effective amount of the ΙΤΝβ analog peptide may be between about 10 mg/kg of body weight and about 20 mg/kg of body weight. In some exemplary embodiments, administering the effective amount of the ΙΤΝβ analog peptide may include intravenously injecting the ΠΤΝΓβ analog peptide.

EXAMPLES

[0048] EXAMPLE 1; IDENTIFYING AN IFNB ANALOG PEPTIDE

[0049] In this example, an exemplary ΠΤΝΓβ analog peptide was identified. At first, a highly conserved peptide in the amino acid sequence of ΠΤΝΓβ was identified by identifying conserved residues in the amino acid sequence of IFN-β, predicting functional residues of IFN-β, determining a mode of interaction of the ΠΤΝΓβ-IFNARl complex, and identifying functionally interacting residues of ΙΤΝβ and IFNARl .

[0050] In order to identify the highly conserved peptide in the amino acid sequence of ΠΤΝΓβ, amino acid sequences of ΠΤΝΓβ in human, mouse, and rat were obtained from UniProt databank. Subsequently, multiple sequence alignment (MSA) was performed using Clustal-W program and the aligned sequences were visualized using the Jalview program. Based on the results of sequence alignment, a peptide including amino acid residues at positions from 83 to 109 of the ΠΤΝΓβ was considered as a highly conserved peptide with an amino acid sequence as set forth in SEQ ID No. 4. The highly conserved peptide included 27 amino acids. [0051] Also, the functional residues of ΠΤΝΓβ involved in ΠΤΝΓβ activity were predicted by identifying highly conserved patches of three-dimensional (3D) structure of the IFNP using ConSurf server. FIG. 2A shows a representation of ΠΤΝΓβ structure with cross-species conservation scores, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 2A, darker colors/shades indicate more conserved residues, so it may be concluded that highly conserved peptide 200 with the amino acid sequence as set forth in SEQ ID No. 4 included several functionally and structurally conserved residues that were expected to have a central role in interaction of the ΠΤΝΓβ and the IFNAR1.

[0052] The mode of interaction of the human ΠΤΝΓβ -IFNARl complex was determined by evaluating the mode of interaction for the murine IfnP-Ifnarl complex and identifying the structurally conserved residues between the human and murine proteins. Moreover, the human IFNP and murine Ιίηβ were structurally aligned using SuperPose and TM-align servers. A structural alignment was also carried out between human IFNAR1 and murine Ifnarl.

[0053] FIG. 2B shows a representation of the highly conserved region in the ΠΤΝΓβ structure, consistent with one or more exemplary embodiments of the present disclosure. The highly conserved peptide is shown in black color. Referring to FIG. 2B, the main IFNAR-binding site of ΠΤΝΓβ encompasses a side of ΠΤΝΓβ that is built up by a-helices 4, 5 and 6, and two loop spacers 202.

[0054] Furthermore, molecular docking was conducted for determining the functionally interacting residues including a ligand-binding site of human IFNAR1 and an IFNAR-binding site of human ΠΤΝΓβ utilizing ClusPro and HADDOCK web tools. Consequently, both blind and high-resolution docking runs were separately performed, and the protein complexes with lowest binding energy were chosen and visualized using LigPlot+ software to understand the functionally interacting residues of ΠΤΝΓβ and IFNARl .

[0055] FIG. 2C shows a human HTN^-IFNARl complex after molecular docking, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 2C, the residues of the highly conserved peptide were apparently involved in the binding of ΠΤΝΓβ 204 to IFNARl.

[0056] In the next step, the library of tolerated peptides was generated by substituting the amino acid in the highly conserved sequence. The library of tolerated peptides was generated using Rosetta backrub with sequence tolerance protocols implemented in the Rosetta3.5 software based on IFNARl -binding site of the IFNP and the most tolerated peptide was determined among hundreds of generated peptides.

[0057] Physicochemical properties of the highly conserved peptide were improved via several amino acid substitutions. Physicochemical features such as theoretical pi, net charge at pH 7, grand average of hydropathicity (GRAVY), half-life, and cell-penetrating capability were calculated using ProtParam web tool, Innovagen web tool, and CellPPD web tool. Critical functional residues involved in interaction with IFNARl and in the biological activity of the IFNP were not substituted. Calculation of the physicochemical properties of the highly conserved peptide with SEQ ID No. 4 revealed some undesirable features. Thus, several properties of the highly conserved peptide were improved using some logical amino acid substitutions to derive the IFNP analog peptide with SEQ ID No. 1 and the IFNP analog peptide with SEQ ID No. 2.

[0058] In the last step, the IFNP analog peptide was selected by improving binding affinity and stability of the tolerated peptides. At first, candidate peptides were selected by improving binding affinity of the tolerated peptide via docking the tolerated peptide with IFNARl. Afterward, the most stable candidate peptide as the IFNP analog peptide was selected by calculating free energy of the candidate peptides. The IFNP analog peptide included the amino acid substitutions of I5F, F6R, I8G, F9W, R10H, D12N, S 13T, S 14L, S 15N, W18L, N19R, E20W, T21V, I22G, and V23F in the amino acid sequence of the IFNp. The IFNp analog peptide had an amino acid sequence as set forth in SEQ ID No. 1 with 27 amino acids.

[0059] After selecting the IFNP analog peptide, a high-resolution docking was conducted based on three-dimensional structures of peptides, predominant binding sites, and computational docking information. FIG. 3A shows interaction mode between highly conserved peptide 302 and IFNARl 300 after a high -resolution peptide-protein docking, consistent with one or more exemplary embodiments of the present disclosure. FIG. 3B shows interaction mode between IFNP analog peptide 304 and IFNARl 300 after a high-resolution peptide-protein docking, consistent with one or more exemplary embodiments of the present disclosure.

[0060] Referring to FIGs. 3A and 3B, IFNP analog peptide 304, similar to highly conserved peptide 302, could specifically bind to the ligand-binding site of IFNARl 300. Regarding the fact that highly conserved peptide 302 of IFNP may activate the IFNAR signaling through interaction with IFNARl 300, independent of IFNAR2, IFNP analog peptide 304 may be capable of inducing downstream signaling through binding to IFNARl 300. Due to its higher coil structure content, IFNP analog peptide 304 may be flexible enough to move readily on the receptor surface, and thereby, form a stable peptide-receptor complex.

[0061] Also, polar and nonpolar contacts of the complexes were determined. The PRODIGY web tool calculates a number of interfacial contacts (ICs) and percentage of the non-interacting surface (NIS) in a protein-protein complex which has been shown to be crucial in physical protein-protein interactions.

[0062] TABLE. 1: Number of interfacial contacts (ICs) in the complex of different peptides with IFNARl

Type of the IC Highly conserved peptide- IFNp analog peptide- IFNARl complexes IFNAR1 complexes

Charged-Charged 4 5

Charged-Polar 5 8

Charged-Apolar 11 10

Polar-Polar 2 9

Polar-Apolar 8 19

Apolar-Apolar 19 10

Total 49 61

[0063] Referring again to TABLE. 1, the number of interfacial contacts (ICs), particularly polar-apolar and charged-polar contacts, between the IFNP analog peptide and IFNARl was substantially higher than the highly conserved peptide and IFNARl. A total number of connections in the IFNP analog peptide-IFNARl complex (61 contacts) were obviously higher than the highly conserved peptide-IFNARl complex (49 contacts) indicating that the IFNP analog peptide with SEQ ID No. 1 may probably form a stable interaction compared to the highly conserved peptide.

[0064] EXAMPLE 2: INTERACTION STABILITY OF THE IFNB ANALOG PEPTIDE

[0065] In this example, interaction stability of the IFNP analog peptide with SEQ ID No. 2 was evaluated using molecular dynamics (MD) and energetic analysis. MD simulations were performed using GROMACS package. At first, three-dimensional structure of the highly conserved peptide with SEQ ID No. 4 and IFNP analog peptide with SEQ ID No. 2 in complex with IFNARl were solvated in a solvation box with 10.5 A distance between the edges of the box and the protein fragments. Na + and CI " ions were added to the box to neutralize the system. Also, SYR6 peptide as a known ΠΤΝΓβ analog peptide was used as a positive control.

[0066] Subsequently, the entire systems were minimized and equilibrated for 100 ps using canonical (NVT) and the isothermal-isobaric (NPT) ensembles. Then, the systems were subjected to a 30 ns MD simulation using the leap-frog algorithm with an integration time step of 0.002 ps. Stability and conformational changes of the ΠΤΝΓβ analog peptide-IFNARl complex as well as the IFNP-IFNARl complex were assessed using plotted root mean square deviation (RMSD) of the backbone atoms and radius of gyration (Rg).

[0067] FIG. 4A shows a plotted root mean square deviation (RMSD) plot for the complex of different peptides with IFNAR1, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 4A, very small, RMSD values, about 0.25 nm at the first 15 nanoseconds (ns) and about 0.3 nm in the second 15-ns of MD simulations, and invariable RMSD values of the ΠΤΝΓβ analog peptide-IFNARl complex 404 imply a high stability and low conformational freedom of the peptide-receptor complex during MD simulations. In contrast, RMSD of SYR6-IFNAR1 complex 402 exhibited some structural fluctuations upon binding to IFNAR1 that may be attributed to unstable structure of SYR6-IFNAR1 complex 402.

[0068] FIG. 4B shows a radius of gyration (Rg) plot for the complex of different peptides with IFNAR1, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 4B, the Rg value of ΠΤΝΓβ-IFNARl complex 406 increased to about 2.3 nm, then decreased to stabilized at about 2.25 nm. The Rg value of SYR6-IFNAR1 complex 408 exhibited a sharp increase from about 2 nm to about 2.05 nm at 20 ns and then stabilizes around this value. The Rg of the IFNP analog peptide-IFNARl complex 410 remained approximately 1.95 nm over the 30 ns MD simulations except for a short period.

[0069] EXAMPLE 3: BINDING AFFINITY OF THE IFNB ANALOG PEPTIDE

[0070] In this example, binding affinity of the highly conserved peptide with SEQ ID No. 4 and the IFNp peptide with SEQ ID No. 2 (MSPEP27) to IFNAR1 was investigated using molecular docking and energetic analysis. At first, an average structure was prepared for each peptide-IFNARl complexes from the last 3 ns MD simulations and subjected to the PRODIGY web tool for calculating the AG of interactions.

[0071] Also, polar and nonpolar contacts of the complexes were determined. The PRODIGY web tool calculates the number of interfacial contacts (ICs) and the percentage of the non- interacting surface (NIS) in a protein-protein complex which has been shown to be crucial in physical protein-protein interactions. In parallel, electrostatic energies were calculated using the PBEQ-Solver tool utilizing 31 snapshot configurations taken from the last 3 nanoseconds (ns) MD productions of the peptide-IFNARl complexes and subsequently averaging the values. The van der Waals (VdW) energies were calculated using a standard GROMACS utility.

[0072] TABLE. 2: Binding energies between different peptides and IFNAR1

Interaction Free Energy Components in complex with IFNAR1 (kcal/mol)

Energy Highly conserved peptide IFNp analog peptide IFNP

ClusPro score -1801.2 -2447.2 -2810.4

HADDOCK score -78.0 + 8.9 -147.3 + 3.1 -162.0 + 5.1

Cluster size 34 28 47

AG interaction -9.7 -10.8 -15.0

ΔΕ electrostatic -175.9 + 25.5 -313.8 + 29.8 -625.7 + 22.1

ΔΕ VdW -59.4 + 6.9 -57.9 + 2.1 -86.3 + 9.6

AG desolvation -9.6 + 9.9 -5.1 + 4.4 41.8 + 9.3

Kd (M) 5.9e-04 6.5e-08 9.2e-12

Non-Interacting Surface (NIS) per property

NIS charged% 28.54 26.21 33.31

NIS apolar% 38.42 29.65 30.82

[0073] Referring to TABLE. 2, the IFNP analog peptide may be most likely capable of binding to IFNAR1, with approximately similar binding energy compared to ΠΤΝΓβ. Also, the highly conserved peptide appears to have less IFNAR1 binding affinity than the IFNP analog peptide due to lower Kd value. Moreover, calculation of non-interacting surfaces implicated important role of electrostatic energies in the interaction selectivity of the peptides with IFNAR1. Therefore, the IFNP analog peptide has higher interaction selectivity with IFNAR1 in comparison with the highly conserved peptide because the IFNP analog peptide has lower percentage of charged non-interacting surface (NIS) and apolar NIS.

[0074] EXAMPLE 4: VALIDATION OF PEPTIDE-RECEPTOR INTERACTION

[0075] In this example, validation of the IFNP analog peptide-IFNARl complex as a peptide- receptor interaction was carried out using intrinsic fluorescence measurements. The IFNP analog peptide had an amino acid sequence as set forth in SEQ ID No. 2. Fluorescence measurements could provide useful information including the binding of molecules to proteins at the molecular level. Intrinsic fluorescence of IFNAR1 would become significantly weakened if physicochemical conditions of local surroundings of IFNAR1 are changed slightly. For example, some factors such as biomolecule binding could be responsible for the weakening of the intrinsic fluorescence of IFNAR1.

[0076] In order to determine the quenching mechanism of the interaction between IFNAR1 and the IFNP analog peptide, the fluorescence studies performed using a spectrofluorimeter with a cell compartment thermostated at 293 K. Protein intrinsic fluorescence experiments were carried out at constant IFNAR1 concentration titrated with different concentrations of IFNP analog peptide to reach a maximum molar ratio of the IFNP analog peptide-IFNARl, and the complex maximum molar ratio of the IFNP analog peptide-IFNARl was 10.

[0077] The concentration of IFNAR1 was 3 μΜ and the concentrations of the IFNP analog peptide were from 0 to 30 μΜ in 50 mM phosphate buffer and pH 7.4 at 298 K. Emission spectra were recorded from 310 nm to 450 nm with an excitation wavelength of about 280 nm and at a scan rate of about 60 nm/min. The results of all fluorescence measurements were corrected by the results of the same titrations of medium without IFNAR1. Spectral resolution was 5 nm for both excitation and emission.

[0078] In these experiments, 1.0 ml solution with a fixed concentration of the IFNP analog peptide was precisely added into the quartz cell with a 1 cm path length and was manually titrated by successive additions of the IFNP analog peptide at 5 min time intervals. The fluorescence emission spectra were then measured, and the maximum fluorescence intensity at a wavelength of about 340 nm was used to calculate the binding constant parameters.

[0079] FIG. 5A shows fluorescence emission spectra of IFNAR1 at different concentrations of the IFNP analog peptide, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 5A, the IFNAR1 fluorescence intensity quenched remarkably as the IFNP analog peptide concentrations increased. This trend indicated the IFNP analog peptide significantly interacts with IFNAR1 in a dose-dependent manner. The reduction in fluorescence intensity resulted from the diminution of the fluorescence quantum yield, which in turn was the result of a lowering of the electronic density after the IFNP analog peptide interacted with the IFNAR1 as the receptor. This interaction caused the changes in the microenvironment of aromatic fluorophores of the IFNAR1. [0080] Fluorescence quenching data from the interaction of the IFNP analog peptide and IFNARl were analyzed to obtain various binding parameters. The binding constant (K a ) and the number of binding (n) were calculated according to the Eq. 1, where Fo is the fluorescence emission intensities of IFNARl in the absence of the IFNP analog peptide, F is the fluorescence emission intensities of IFNARl in the presence of the IFNP analog peptide, [P] is the IFNP analog peptide concentration, K a is the binding constant, and n is the apparent molar ratio of [IFNp analog peptide]/ [IFNARl] complex.

Log^ = LogK a + nLog[P] Eq. 1

[0081] FIG. 5B shows a plot for quenching of the IFNARl intrinsic fluorescent by the IFNP analog peptide, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 5B, the slope of the straight- line equals to n as the number of bindings, and the intercept on Y-axis equal to log K a . The values of Ka and n at 298 K were 3.67(+ 0.24) x 10 7 M "1 and 1.17(+ 0.09), respectively. The "n" and K a values indicated that IFNARl significantly interacts with the peptide and forms equimolar complex. It is evident from K a value that the binding of the IFNP analog peptide to the IFNARl is enough strong to make a peptide-receptor inclusion complex.

[0082] EXAMPLE 5: THERAPEUTIC EFFECTS OF THE IFNB ANALOG PEPTIDE

[0083] In this example, therapeutic effects of the IFNP analog peptide with SEQ ID No. 1 on the development of progressive experimental autoimmune encephalomyelitis (EAE) were studied. At first, EAE was induced in 8 weeks old C57BL/6 male mice with a weight between 20 grams and 25 grams. In order to induce EAE, mice were anesthetized with isoflurane and subcutaneously injected with 100 μΐ of MOG (myelin oligodendrocyte glycoprotein) with a concentration of about 2.5 g/1 suspended in 100 μΐ complete Freund's adjuvant (CFA) into the hind flank. Mice were also intraperitoneally injected with 400 ng of pertussis toxin in 200 μΐ of phosphate buffered saline (PBS). A second, identical injection of pertussis toxin was given two days following the first immunization. Injections were distributed over four spots, for example about 100 μΐ per site across the flank areas.

[0084] In order to investigate whether treatment with the IFNP analog peptide could modulate the EAE, seven experimental groups were designed. One hundred and twenty-six mice were divided randomly into seven groups: (i) a vehicle group with 18 mice received injection of PBS solution as vehicle at day 14 of EAE induction, (ii) sham group with 18 mice received pertussis toxin without MOG; (iii) EAE groups with 90 mice received pertussis toxin with MOG and were divided into five separate subgroups (18 mice per subgroup), (iiia) without vehicle, the IFNP analog peptide and IFNP injection, (iiib) with 10 mg/kg body weight injection of the IFNP analog peptide at day 14 of EAE induction (iiic) with 20 mg/kg body weight injection of the IFNP analog peptide at day 14 of EAE induction (iiid) with 10 mg/kg body weight injection of IFNP at day 14 of EAE induction (iiie) with 20 mg/kg body weight injection of IFNP at day 14 of EAE induction.

[0085] In order to bypass the blood-brain barrier and other mechanisms that limit systemic drug distribution into the brain, intracerebroventricular injection (ICVI) route of administration was selected to allow CNS entrance of exact concentrations used for both IFNP analog peptide and IFNp. The immunized mice received intravenous injections of 100 μΐ containing IFNP and the IFNP analog peptide with doses of 0, 10 and 20 mg/kg dissolved in 0.2 M PBS into the right lateral ventricle of the mouse on day 14 of EAE induction.

[0086] After that, in order to conduct clinical evaluation, mice of the experimental groups were blinded scored as follows: score 0 for no overt signs of disease, score 1 for limp tail, score 2 for limp tail and hind limb weakness, score 3 for partial hind limb paralysis of one side (hemiparesis) or both sides (paraparesis), score 4 for complete hind limb paralysis of one side (hemiplegia) or both sides (paraplegia), score 5 for moribund state. The day of immunization was considered as EAE day 0 and clinical behavior of mice was scored daily. Mice were typically observed for 3 weeks, during which mice remained chronically paralyzed with the onset of paralysis between 12 and 16 days after immunization and a maximum score of 3 to 4 in most mice.

[0087] FIG. 6 shows mean clinical scores of different experimental groups after immunization, consistent with one or more exemplary embodiments of the present disclosure. The experimental groups were sham group treated with pertussis toxin without MOG 600, EAE- induced mice without any injection 602, EAE-induced mice treated with PBS solution as a vehicle group 604, EAE-induced mouse treated with 10 mg/kg IFNP analog peptide 606, EAE- induced mice treated with 20 mg/kg IFNP analog peptide 608, EAE-induced mice treated with 10 mg/kg IFNP 610, EAE-induced mice treated with 20 mg/kg IFNP 612. All of the treatments were done on day 14 post-immunization.

[0088] Referring to FIG. 6, comparison area under curve (AUC) between IFNP analog pep tide-treated groups 606 and 608 and control groups of vehicle group 604 and EAE group 602 revealed that the IFNP analog peptide-treated groups 606 and 608 showed a higher decrease in mean clinical score, and IFNP analog peptide treatment significantly attenuated severity of clinical EAE (P < 0.05 and P < 0.01). Therefore, it may be concluded that IFNP analog peptide had a modulatory effect on EAE progression. The IFNP analog peptide could efficiently bind to IFNAR1 and suppress neuroinflammation in-vivo. Also, IFNP analog peptide had protective effects against MOG-induced EAE via reduction of immune dysfunction and inflammation.

[0089] Referring again to FIG. 6, as expected, IFNP-treated groups 610 and 612 showed a significant reduction in the clinical score in comparison with control groups of vehicle group 604 and EAE group 602. Comparison between IFNP analog peptide-treated groups 606 and 608 and IFNP-treated groups 610 and 612 showed that there was no statistically significant difference between effects of IFNP analog peptide and IFNP on changes in clinical scores during treatments, and a significant amelioration in clinical severity was observed for both IFNP-treated groups and IFNP analog peptide-treated groups in a time-dependent manner.

[0090] Referring again to FIG. 6, comparison between EAE-induced mouse treated with 10 mg/kg IFNP analog peptide 606 and EAE-induced mice treated with 20 mg/kg IFNP analog peptide 608 indicates that raising the dose of the IFNP analog peptide could further protect mice from EAE clinical symptoms. In addition, an increased in drug dosage for both IFNP and IFNP analog peptide improved disease protection with the same pattern.

[0091] EXAMPLE 6: EFFECTS OF THE IFNB ANALOG PEPTIDE ON INFLAMMATION MEDIATORS

[0092] In this example, effects of the IFNP analog peptide as the IFNP analog peptide on the expression of some related inflammations mediators such as matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP9) were studied using quantitative real-time polymerase chain reaction (PCR).

[0093] FIG. 7 A shows an expression fold change of matrix metalloproteinase 2 (MMP2) in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure. FIG. 7B shows an expression fold change of matrix metalloproteinase 9 (MMP9) and MMP2 in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure. The EAE mice were treated with PBS as the vehicle group, and dosage of the IFNP analog peptide and IFNP was 20 mg/kg in IFNP analog peptide-treated mice and IFNP-treated mice groups. After that RNA of the cells was extracted and their cDNA was synthesized. [0094] Referring to FIG. 7A and 7B, treatment with IFNP analog peptide and IFNP showed a significant decrease in MMP2 and MMP9 gene expression fold change in the brain tissue compared to the vehicle group (P < 0.05). 0.05). However, reduction of these inflammation mediators was more in IFNP-treated mice.

[0095] Moreover, after 21 days of EAE induction, mice were euthanized and their blood was obtained via cardiac puncture and plasma was prepared. Expression levels of tumor necrosis factor- alpha (TNFa) and interleukin ip (ILip) as circulating inflammatory biomarkers were measured in the plasma using quantitative enzyme-linked immunosorbent assay (ELISA) kits.

[0096] FIG. 7C shows a concentration change of tumor necrosis factor alpha (TNFa) in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure. FIG. 7D shows a concentration change of interleukin 1 beta (ILip) in IFNP analog peptide-treated mice and IFNP-treated mice, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIGs. 7C and 7D, treatment with IFNP analog peptide and IFNP showed a significant decrease in plasma levels of TNFa and ILip compared to the vehicle group (P-value < 0.05). However, reduction of these inflammation mediators was more in IFNP-treated mice.

[0097] Furthermore, brain inflammatory response after IFNP analog peptide treatment as the IFNP analog peptide was evaluated by measuring the number of IL-17 positive cells, CD l ib positive cells, and CD45 positive cells. IL-17 is the most important pro-inflammatory cytokine produced by T-helper cells, CD l ib is an activated macrophages and microglia marker, and CD45 is a general leukocytes marker.

[0098] After 21 days of EAE induction, mice were euthanized and were transcardially perfused with PBS. Then, the brains were dissected; cerebral cortex was isolated and snap-frozen in liquid nitrogen. FIG. 8A shows a percentage of interleukin 17 (IL17) positive cells, cluster of differentiation l ib (CD l ib) positive cells, and cluster of differentiation 45 (CD45) positive cells in each μπι2 of mice cerebral cortex after injection of an IFNP analog peptide, consistent with one or more exemplary embodiments of the present disclosure.

[0099] Referring to FIG. 8A, there is a significant reduction of IL-17, CD l ib, and CD45 in the cerebral cortex after IFNP analog peptide treatment, particularly in the higher dosage, for example, 20 mg/kg. Quantitative analysis revealed that this reduction occurs in a dose- dependent manner. The percentage of IL-17, CD 1 lb and CD45 positive cells in the mice treated with 20 mg/kg of IFNP analog peptide was all less than half of the untreated EAE mice, and the most significant decrease was observed for CD45, especially when 20 mg/kg of IFNP analog peptide was applied.

[00100] FIG. 8B shows a percentage of IL17, CDl lb and CD45 positive cells in each μιη 2 of mice cerebral cortex after IFNP injection, consistent with one or more exemplary embodiments of the present disclosure. Referring to FIG. 8B, the percentage of IL-17, CDl lb, and CD45 positive cells were considerably decreased in mice treated with IFNP, particularly in the 20 mg/kg dosage.

[00101] Referring again to FIGs. 8A and 8B, there was no statistically significant difference between effects of the IFNP analog peptide and IFNP on the expression of IL-17, CDl lb, and CD45 during treatments. In-vivo analyses revealed that the IFNP analog peptide could lessen inflammation of the EAE mice by modulating the levels of inflammatory mediators.

INDUSTRIAL APPLICABILITY

[00102] Applicants have found that the exemplary interferon-beta (IFNP) analog peptide of the present disclosure is particularly suited for industrial applications. By way of example, industrial applications may include pharmaceutical industry. The exemplary interferon-beta (IFNP) analog peptide may be used as a therapeutic formulation for treating multiple sclerosis (MS) or viral infections in a patient with low side effects.

[00103] While the foregoing has described what are considered to be the best mode and/or other examples, it is understood that various modifications may be made therein and that the subject matter disclosed herein may be implemented in various forms and examples, and that the teachings may be applied in numerous applications, only some of which have been described herein. It is intended by the following claims to claim any and all applications, modifications and variations that fall within the true scope of the present teachings.

[00104] Unless otherwise stated, all measurements, values, ratings, positions, magnitudes, sizes, and other specifications that are set forth in this specification, including in the claims that follow, are approximate, not exact. They are intended to have a reasonable range that is consistent with the functions to which they relate and with what is customary in the art to which they pertain.

[00105] The scope of protection is limited solely by the claims that now follow. That scope is intended and should be interpreted to be as broad as is consistent with the ordinary meaning of the language that is used in the claims when interpreted in light of this specification and the prosecution history that follows and to encompass all structural and functional equivalents. Notwithstanding, none of the claims are intended to embrace subject matter that fails to satisfy the requirement of Sections 101, 102, or 103 of the Patent Act, nor should they be interpreted in such a way. Any unintended embracement of such subject matter is hereby disclaimed.

[00106] Except as stated immediately above, nothing that has been stated or illustrated is intended or should be interpreted to cause a dedication of any component, step, feature, object, benefit, advantage, or equivalent to the public, regardless of whether it is or is not recited in the claims.

[00107] It will be understood that the terms and expressions used herein have the ordinary meaning as is accorded to such terms and expressions with respect to their corresponding respective areas of inquiry and study except where specific meanings have otherwise been set forth herein. Relational terms such as first and second and the like may be used solely to distinguish one entity or action from another without necessarily requiring or implying any actual such relationship or order between such entities or actions. The terms "comprises," "comprising," or any other variation thereof, are intended to cover a non-exclusive inclusion, such that a process, method, article, or apparatus that comprises a list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, article, or apparatus. An element proceeded by "a" or "an" does not, without further constraints, preclude the existence of additional identical elements in the process, method, article, or apparatus that comprises the element.

[00108] The Abstract of the Disclosure is provided to allow the reader to quickly ascertain the nature of the technical disclosure. It is submitted with the understanding that it will not be used to interpret or limit the scope or meaning of the claims. In addition, in the foregoing Detailed Description, it can be seen that various features are grouped together in various implementations. This is for purposes of streamlining the disclosure, and is not to be interpreted as reflecting an intention that the claimed implementations require more features than are expressly recited in each claim. Rather, as the following claims reflect, inventive subject matter lies in less than all features of a single disclosed implementation. Thus, the following claims are hereby incorporated into the Detailed Description, with each claim standing on its own as a separately claimed subject matter.

[00109] While various implementations have been described, the description is intended to be exemplary, rather than limiting and it will be apparent to those of ordinary skill in the art that many more implementations and implementations are possible that are within the scope of the implementations. Although many possible combinations of features are shown in the accompanying figures and discussed in this detailed description, many other combinations of the disclosed features are possible. Any feature of any implementation may be used in combination with or substituted for any other feature or element in any other implementation unless specifically restricted. Therefore, it will be understood that any of the features shown and/or discussed in the present disclosure may be implemented together in any suitable combination. Accordingly, the implementations are not to be restricted except in light of the attached claims and their equivalents. Also, various modifications and changes may be made within the scope of the attached claims.