Peptides and their use as antiviral agents

文档序号:1408820 发布日期:2020-03-06 浏览:31次 中文

阅读说明:本技术 肽及其用作抗病毒剂的用途 (Peptides and their use as antiviral agents ) 是由 ***·拉吉克·***·*** 于 2017-05-26 设计创作,主要内容包括:本文公开了具有抗病毒特性的肽。所述肽包括右旋糖(D)或右旋/左旋(L)氨基酸的混合物,具有针对多种类型的包膜病毒和基质蛋白层病毒的抗病毒特性,可用作治疗或预防包膜病毒和基质蛋白层病毒感染的药物组合物。(Disclosed herein are peptides having antiviral properties. The peptides comprise a mixture of dextrose (D) or D/L (L) amino acids, have antiviral properties against a variety of types of enveloped and matrix protein layer viruses, and are useful as pharmaceutical compositions for treating or preventing infection by enveloped and matrix protein layer viruses.)

1. Use of a therapeutically effective amount of a peptide, or a pharmaceutical composition of any of said peptides, in the manufacture of a medicament for treating an infection caused at least in part by a matrix protein layer virus or an enveloped virus in a host animal, wherein the virus is not an influenza virus or an orthomyxovirus, the peptide comprising the sequence twftfin or an analogue or derivative of twftfin of the formula a1-a2-A3-a4-A5-A6-a7, wherein a1 is t or w, a2 is k or w, A3 is s or f, a4 is r or t, A5 is f, h, i, n or w, A6 is i or D, a7 is n or a, and wherein any one or any two of a1, a2, A3, a4, A5, A6 or a7 is optionally substituted by another D-amino acid or L-amino acid.

2. The use of claim 1, wherein the virus is a retrovirus, lentivirus, paramyxovirus, flavivirus, hepatitis c virus, tick-borne encephalitis, yellow fever, dengue virus, filovirus, togavirus, bunyavirus, herpes virus, hepnavirus, paramyxovirus, or coronavirus.

3. The use of claim 1, wherein the virus is HIV, ebola, marburg, rubella, hantavirus, arenavirus, cytomegalovirus, hepatitis b virus, or circovirus.

4. The use of claim 1, wherein the infection is AIDS, herpes or SARS.

5. The use according to any one of claims 1 to 4, wherein the peptide comprises the sequence twftfin, X/xwftffin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftX/xn, twftfX/xn, twftfiX/X or X/xwftfiX/X, wherein X/X is glycine, or any D-amino acid, or any L-amino acid, and wherein in each sequence one other amino acid is inserted, deleted or changed to a different D-amino acid, or glycine, or to any L-amino acid by one amino acid.

6. Use according to any one of claims 1 to 4, wherein the peptide comprises the sequence X/xwftfiX/X.

7. The use according to any one of claims 1 to 4, wherein the peptide comprises the sequence PSP IV A17, PSP V A19 or PSP VI A1.

8. The use according to any one of claims 1-4, wherein the peptide further comprises an N-terminal and/or C-terminal cysteine residue.

9. The use of any one of claims 1-4, wherein the peptide further comprises one or more solubility tags, each of which is independently selected.

10. Use according to any one of the preceding claims, wherein the peptide is in cyclic form.

11. Use according to any one of the preceding claims, wherein the peptide has a viral IC50Less than 100. mu.M.

12. Use according to any one of the preceding claims, wherein the medicament is suitable for nasal, pulmonary, oral or parenteral administration.

13. Use according to any one of the preceding claims, wherein the medicament is for prophylactic treatment.

14. A kit comprising a therapeutically effective amount of a peptide comprising the sequence twftfin or an analog or derivative of twftfin, or a pharmaceutical composition of any of the peptides, wherein the virus is not an influenza virus or an orthomyxovirus, or instructions for treating an infection in a host animal caused at least in part by a matrix protein layer virus or an enveloped virus.

Technical Field

The present invention relates to therapeutic peptides having broad spectrum antiviral activity against a variety of enveloped and matrix protein layer viruses.

Background

Viral infection by enveloped and matrix protein layer viruses remains a serious problem in humans. Although influenza viruses have attracted the attention of many researchers, viral therapy of other enveloped and matrix protein layer viruses, including but not limited to herpes simplex virus, cytomegalovirus, respiratory syncytial virus and human immunodeficiency virus, remains a significant problem to be solved in the medical field.

Respiratory Syncytial Virus (RSV) belongs to the Paramyxoviridae family (Paramyxoviridae) and comprises an enveloped, negative-sense, single-stranded, non-RNA genome1. Although RSV is one of the important pediatric pathogens, they can affect individuals of various ages, especially elderly and immunocompromised individuals. RSV only infects humans and is highly contagious. Currently, there is no approved therapy available for treatment2. It is estimated that about 3400 ten thousand children under 5 years of age are infected worldwide each year, resulting in about 400 ten thousand hospitalized patients and 10 to 20 ten thousand deaths. Typically, RSV causes upper respiratory tract infections, but 20-30% of infected children exhibit symptoms of lower respiratory tract infections, such as bronchiolitis and/or pneumonia. Symptoms of RSV infection include weakness, irritability, cough, wheezing, crackles, shortness of breath, and loss of appetite. Factors such as heart disease, chronic lung disease in premature infants, immunodeficiency and immunosuppression can increase and exacerbate the symptoms of the disease. The current treatment method for RSV infection mainly aims at the preventive use of patients under high-risk or extremely high-risk conditions3. There is a need for more general therapies as well as post-infection therapies.

Cytomegalovirus (CMV) belongs to the family of Herpesviridae (Herpesviridae), which comprises a linear double stranded DNA genome. The viral genome is surrounded by nucleocapsid proteins, which are themselves surrounded by an envelope (matrix layer) and a lipid bilayer4,5. CMV infects people of various ages, and by the age of 40, most people are reportedCMV is infected at some stage in their life. Normally, CMV infection does not show any symptoms, but people with a weaker immune system and infants with congenital infections are more prone to serious complications6,7. Symptoms of CMV infection range from asymptomatic, fever, tiredness to severe symptoms, which may damage internal organs, eyes and brain. Although some antiviral drugs, such as valganciclovir, ganciclovir, cidofovir, foscarnet or combinations thereof, can be used to treat retinitis, these drugs may cause serious side effects and may not cure the infection. There is a need for a more general therapy, and a therapy with fewer or less side effects8-10

Herpes simplex viruses (HSV-1 and HSV-2) belong to the family of Herpesviridae (Herpesviridae) and comprise linear double stranded DNA as the genome. The genome of herpes simplex virus is surrounded by icosahedral nucleocapsid proteins, which are themselves surrounded by a capsule (matrix layer) and a lipid bilayer5,11. These viruses often cause skin, eye, lip and genital infections. Symptoms include painful blisters or ulcers at the site of infection, tingling around the mouth, genitalia and buttocks, swollen lymph nodes, fever and body pain. Herpes simplex virus may also cause meningitis and encephalitis in immunocompromised individuals and neonatal herpes in newborns11. According to the recent estimates of the world health organization, about 35 hundred million people worldwide are infected with HSV-112,13Approximately 5 million people infect HSV-214. Antiviral drugs for herpes simplex virus, such as acyclovir, famciclovir and valacyclovir, are currently used to treat HSV infection, but are generally only effective in alleviating the symptoms of the disease; it does not cure the infection15,16

Human Immunodeficiency Virus (HIV) belongs to the family of Retroviridae (Retroviridae), which contains as its genome two single-stranded RNAs. The viral genome is surrounded by a nucleocapsid protein, which is itself surrounded by a lipid bilayer associated with a matrix protein17. HIV attacks the human immune system and causes acquired immunodeficiency syndrome (AIDS). AIDS patients are more susceptible to weakening of immune systemThere are an unlimited number of infections that can be life threatening. There are currently therapies for treating HIV infection, but none are capable of curing the infection18,19

Despite the high prevalence of viral infections, there are still few approved drugs for the treatment and/or prevention of enveloped virus infections, such as those disclosed herein. In addition, current therapies still suffer from problems with suboptimal efficacy and severe toxicity and are hampered by the high cost of treatment and the emergence of resistant strains.

Pharmaceutical companies face increasing challenges in expanding their product lines, particularly due to non-druggy targets such as protein-protein interactions20. Although protein-protein interactions are part of host-pathogen interactions, protein-protein interactions also play a crucial role in many cellular functions of healthy cells. Modulation of protein-protein interactions is considered an attractive approach to alleviating many diseases, including inhibiting the entry or replication of pathogens21. However, it has been reported that small molecules are generally not effective in inhibiting this interaction22. Although monoclonal antibodies are highly specific and effective in inhibiting or modulating these interactions, they often lack sufficient tissue or cell permeability23. Without being bound by theory, it is believed herein that a macrocyclic peptide with cell permeability may serve as an optimal solution. Given the high variability of many viruses, drugs targeting protein-protein interactions should have sufficient flexibility to withstand such mutations, and thus macrocyclic peptides may also be the best choice. In addition, peptides have many advantages over small molecules, including high specificity, selectivity, excellent potency, and they are generally well tolerated24,25. Also, due to their lower manufacturing complexity and better target accessibility (target accessibility), peptides may be more favored over other macromolecules such as antibodies24. Without being bound by theory, it is believed herein that peptides may have more advantages and overcome more limitations than small or large molecules24-30

Novel peptide therapeutics (FPT) have been discovered having a broad spectrum of activity against influenza viruses31,32. The mechanism of action of FPT on influenza virus was investigated. Without being bound by theory, it is believed herein that FPT exerts its effects by binding to and altering the integrity of the matrix protein (M1) of influenza virus. Later, it was found that influenza virus matrix proteins have sufficient structural similarity to HIV, HSV, RSV and CMV, and therefore FPT is also active against these viruses as well as other enveloped and matrix protein layer viruses.

Disclosure of Invention

It was surprisingly found that a peptide comprising the sequence twftfin is active against a variety of enveloped and matrix proteoliposviruses. It has also been surprisingly found that peptides comprising sequence variants of twftfin, in which one or two other amino acids are independently inserted, deleted or changed to a different D-amino acid, glycine or any L-amino acid, also have high activity against a variety of enveloped and matrix proteoliposviruses. The peptides described herein typically exhibit many times greater antiviral activity than peptides comprising the entire L-amino acid sequence, and exhibit a broad spectrum of activity against a variety of enveloped and matrix proteoliposviruses.

In an exemplary embodiment of the invention, described herein are peptides comprising dextrorotatory amino acids or mixtures of dextrorotatory/levorotatory amino acids, also referred to herein as FTP. In another embodiment, described herein are peptides having antiviral properties against enveloped viruses or matrix protein layer viruses. In another embodiment, described herein are peptides having antiviral properties against a variety of enveloped and matrix protein layer viruses. In another exemplary embodiment, use of any one or more of the peptides described herein in the manufacture of a medicament for treating or preventing infection by enveloped and matrix protein layer viruses is described. In another exemplary embodiment, pharmaceutical compositions, dosage forms and dosage units comprising any one or more of the peptides described herein are described for use in treating or preventing enveloped virus and matrix protein layer virus infections. In another exemplary embodiment, described herein is a method for treating or preventing enveloped and matrix protein layer virus infections, wherein the method comprises administering any one or more of the peptides described herein, or pharmaceutical compositions, dosage forms, dosage units and medicaments comprising the peptides.

In another embodiment, the enveloped virus or matrix protein layer virus is a retrovirus, lentivirus, paramyxovirus, flavivirus, hepatitis c virus, tick-borne encephalitis, yellow fever, dengue virus, filovirus, togavirus, bunyavirus, herpesvirus, hepnavirus, paramyxovirus, or coronavirus. In another embodiment, the enveloped virus or matrix protein layer virus is herpes simplex virus, cytomegalovirus, respiratory syncytial virus, or human immunodeficiency virus. In another embodiment, the virus is HIV, ebola, marburg, rubella, hantavirus, arenavirus, cytomegalovirus, hepatitis b virus, or a circovirus (torovirus).

In another embodiment, the infection is AIDS, herpes or SARS.

In another embodiment, described herein are peptides comprising the sequence tksrfX/xn (formula I). In another embodiment, described herein are peptides comprising the sequence tX/xsrfin (formula II). In another embodiment, described herein is a peptide comprising the sequence twX/xrfin (formula V). In another embodiment, described herein are peptides comprising the sequence twfX/xfin (formula VI). In another embodiment, described herein are peptides comprising the sequence X/xwftfin (formula VII). In another embodiment, described herein are peptides comprising the sequence wwftfiX/x (formula VIII). In another embodiment, described herein are peptides comprising the sequence wwftX/xia (formula IX). In another embodiment, described herein are peptides comprising the sequence X/xwftfiX/X (formula X). In each of the foregoing sequences, X/X is glycine, or any D-amino acid, or any L-amino acid.

In another embodiment, any of the peptides described herein further comprises an N-terminal and/or C-terminal cysteine residue. When both the N-and C-termini comprise a cysteine residue, the peptide may be cyclized. It will be appreciated that in each case the cysteine residues are independently selected from D-cysteine and L-cysteine.

In another embodiment, any of the peptides described herein further comprises one or more solubility tags. Illustratively, a solubility tag is included at the N-terminus and/or the C-terminus. It is to be understood that the peptides described herein (including cyclic derivatives thereof) comprising N-terminal and/or C-terminal cysteine residues may also comprise one or more solubility tags. It is also understood that in each case, each solubility tag is independently selected.

Additional details regarding FPT are disclosed in PCT International application Ser. No. PCT/IB2016/057146, the entire disclosure of which is incorporated herein by reference.

In another embodiment, analogs and derivatives of the peptides described herein are described. It is to be understood that the analogs and derivatives described herein do not include the peptides disclosed in PCT international publication No. WO 2009/151313. In another embodiment, the peptides described herein do not include any of the sequences disclosed in PCT international publication No. WO 2009/151313.

As used herein, the term isolated peptide generally refers to a peptide in vitro and/or a peptide in bulk form (peptide in bulk form). It is to be understood that the isolated peptide may be in the form of a mixture with other compounds, and/or in the form of a solution or suspension.

The sequences of the peptides described herein are typically listed using the conventional one-letter code for each amino acid. The upper case codes correspond to the left-handed form of the amino acid, while the lower case codes correspond to the right-handed form of the amino acid. For example, the single letter code "A" represents L-alanine, and the single letter code "a" represents D-alanine; "R" represents L-arginine and "R" represents D-arginine, etc.

In another embodiment, described herein is an isolated peptide comprising the sequence a1-a2-A3-a4-A5-A6-a7, wherein a1 is t or w, a2 is k or w, A3 is s or f, a4 is r or t, A5 is h, i, n or w, A6 is i or D, a7 is n or a, and any one or two of a1, a2, A3, a4, A5, A6 or a7 is optionally substituted with another D-amino acid, glycine or any L-amino acid. In one variation, described herein are peptides in which one or two of a1, a2, A3, a4, a5, a6, or a7 is replaced with a corresponding L-amino acid.

In another embodiment, described herein is an isolated peptide comprising the sequence a1-a2-A3-a4-A5-A6-a7, wherein a1 is t or w, a2 is k or w, A3 is s or f, a4 is r or t, A5 is h, i, n or w, A6 is i or D, a7 is n or a, and wherein any one of a1, a2, A3, a4, A5, A6 or a7 is optionally substituted with another D-amino acid, glycine or any L-amino acid. In one variation, described herein are peptides in which one of a1, a2, A3, a4, a5, a6, or a7 is replaced with a corresponding L-amino acid.

In another embodiment, described herein is an isolated antiviral peptide comprising a sequence selected from the group consisting of: twftfin, X/xwftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftX/xn, twftfiX/X, and X/xftfix/X.

In another embodiment, described herein is an isolated antiviral peptide comprising a sequence selected from the group consisting of: twftfin, X/xwftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftX/xn, twftfX/xn, twftfiX/X and X/xftfix/X, one additional amino acid being inserted, deleted or changed in each sequence to a different D-amino acid, glycine or any L-amino acid.

In another embodiment, described herein are isolated antiviral peptides comprising a sequence selected from the group consisting of PSP IV a17, PSP VA19, and PSP VI a 1.

In another embodiment, described herein is a peptide comprising a sequence set forth in any one of tables 1-8. It is understood that functional variants of the peptides in these tables that retain biological activity are also within the scope of the invention described herein.

Drawings

FIG. 1: in vitro H-RRKKcwftfiac-OH (PSP VI A1) against RSV A2. The ability of the peptides to inhibit replication of RSV a2 was determined by CPE assay on Hep-2 cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 2: in vitro H-RRKKcwftfiac-OH (PSP IV A17) against RSV A2. The ability of the peptides to inhibit replication of RSV a2 was determined by CPE assay on Hep-2 cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 3: the effect of PSP VI A1 against HSV-2 in vitro. The ability of the peptides to inhibit HSV replication was determined by CPE assay on Vero cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 4: PSP IV A17 is resistant to HSV-2 in vitro. The ability of the peptides to inhibit HSV replication was determined by CPE assay on Vero cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 5: PSP VI a1 was resistant to CMV action in vitro. The ability of the peptide to inhibit CMV replication was determined by CPE assay on HFFF-2 cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 6: PSP IV a17 was resistant to CMV action in vitro. The ability of the peptide to inhibit CMV replication was determined by CPE assay on HFFF-2 cells. Experiments were performed in triplicate and in the presence of DMSO or increasing concentrations of test compound.

FIG. 7: the in vitro PSP VI A1 has anti-HIV-192 UG029 effect. The ability of the peptides to inhibit HIV replication was determined by reverse transcriptase assay on PBMCs. The experiments were performed in DMSO or increasing concentrations of peptide. Results are presented as the average of three measurements.

FIG. 8: anti-HIV-192 UG029 activity of H-cwwftfinc-OH (PSP V A19-C) in vitro. The ability of the peptides to inhibit HIV replication was determined by reverse transcriptase assay on PBMC. The experiments were performed in DMSO or increasing concentrations of peptide. Results are presented as the average of three measurements.

FIG. 9: anti-HIV-192 UG029 activity of H-RRKKtwftfin-OH (PSP IV A17-LS) in vitro. The ability of the peptides to inhibit HIV replication was determined by reverse transcriptase assay on PBMCs. The experiments were performed in DMSO or increasing concentrations of peptide. Results are presented as the average of three measurements.

FIG. 10: cytotoxic effects of antiviral peptides in human hepatocytes. Antiviral peptides H-RRKKctwdrfin-OH (PSP III B3), H-RRKKctwfrfin-OH (PSP III B5), H-RRKKctwsrffin-OH (PSP III B16), H-RRKKctwrrfin-OH (PSP III B19), H-RRKKctwyrfin-OH (PSP III B3)B20) And the cytotoxic effect of H-RRKKctwfffpic-OH (PSP IV A5). Human hepatocytes at two different concentrations HepargTM(cells with a proven differentiated hepatocyte phenotype under cell culture conditions) toxic effects were tested. Data are presented as mean ± standard deviation of three measurements. The experiment was performed as described in example 3.

FIG. 11: cytotoxic effects of antiviral peptides in human hepatocytes. Cytotoxic action of the antiviral peptides PSP IV A17, H-RRKKcwftfinc-OH (PSP V A19), H-RRKKcmwfftfinc-OH (PSP V A11), PSP VI A1 and H-RRKKcwftfisc-OH (PSP VI A16). Human hepatocytes at two different concentrations HepargTM(cells with a demonstrated phenotype of differentiated hepatocytes under cell culture conditions) were tested for toxic effects. Data are presented as mean ± standard deviation of three measurements. The experiment was performed as described in example 3.

FIG. 12: cytotoxic effects of the antiviral peptide PSP V a19 on MDCK and a549 cells. Cytotoxic effect of the antiviral peptide PSP VA 19. The toxic effects of the above peptides on MDCK and a549 cells were tested at a number of different concentrations and compared to untreated controls, as described in example 4. Data are presented as mean ± standard deviation of three measurements.

FIG. 13: cytotoxic effects of the antiviral peptide PSP VI a1 on MDCK and a549 cells. Cytotoxic effects of the antiviral peptide PSP VIA 1. The toxic effect of the above peptides on MDCK and a549 cells was tested at various concentrations and compared to untreated controls as described in example 4. Data are presented as mean ± standard deviation of three measurements.

FIG. 14: hemolytic activity of different antiviral peptides at a concentration of 30. mu.M. The hemolytic activity of triton X-100 or the different antiviral peptides PSP III B5, PSP IV A17, PSP V A19 and PSP VI A1 was tested at a concentration of 30. mu.M as described in example 5 and compared to untreated controls.

Detailed Description

The invention will now be further described by means of the following exemplary embodiments. In the following paragraphs, different exemplary aspects of the present invention will be defined in more detail. Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous. It will be appreciated, however, that the invention is not limited to such feature or features as are indicated as being preferred or advantageous, and that no feature or features as are indicated as being preferred or advantageous is essential.

The practice of the present invention will employ, unless otherwise indicated, conventional techniques of chemistry, biochemistry, molecular biology, recombinant DNA technology, cell biology, immunology and bioinformatics, which are within the skill of the art. Such techniques are explained fully in the literature.

Throughout this disclosure, amino acids are represented using standard one-letter codes according to the IUPAC/IUB guidelines. In the sequences listed herein, the leftmost amino acid at the end of any sequence represents the amino terminus (optionally represented by H-and the rightmost amino acid represents the carboxy terminus (optionally represented by-OH). In addition to achiral glycine, the amino acids in the levorotatory form are indicated herein by upper case letters, while the amino acids in the dextrorotatory form are indicated by lower case letters. It will be understood that reference to an L-amino acid includes glycine unless otherwise indicated.

The present invention relates to peptides having antiviral activity. In addition, the invention relates to peptides having antiviral activity against more than one enveloped or matrix protein layer virus. In addition, the invention relates to peptides having antiviral activity against more than one subtype of enveloped or matrix protein layer virus. In addition, the present invention relates to pharmaceutical compositions comprising the peptides described herein, and methods of preventing and/or treating viral infections using the peptides described herein.

Exemplary embodiments of the present invention are described by the following items:

a sequence of twftfin or an isolated peptide comprising the sequence of twftfin.

An isolated peptide having a sequence that is an analog or derivative of twftfin or a sequence comprising an analog or derivative of twftfin.

An isolated peptide of sequence a1-a2-A3-A4-A5-A6-a7 or comprising sequence a1-a2-A3-A4-A5-A6-a7, wherein a1 is t or w, a2 is k or w, A3 is s or f, A4 is r or t, A5 is f, h, i, n or w, A6 is i or D, a7 is n or a, and wherein any one or any two of a1, a2, A3, A4, A5, A6 or a7 is optionally substituted with another D-amino acid or an L-amino acid.

A peptide of sequence a1-a2-A3-A4-A5-A6-A7 or comprising the sequence a1-a2-A3-A4-A5-A6-A7, wherein a1 is t or w, a2 is k or w, A3 is s or f, A4 is r or t, A5 is h, i, n or w, A6 is i or D, A7 is n or a, and wherein any one or any two of a1, a2, A3, A4, A5, A6 or A7 is optionally substituted with another D-amino acid or an L-amino acid.

The peptide of the preceding item, wherein one or two of a1, a2, A3, a4, a5, a6, or a7 is replaced with a corresponding L-amino acid.

An isolated peptide of sequence a1-a2-A3-A4-A5-A6-a7 or comprising sequence a1-a2-A3-A4-A5-A6-a7, wherein a1 is t or w, a2 is k or w, A3 is s or f, A4 is r or t, A5 is i or w, A6 is i or D, a7 is n or a, and wherein any one or any two of a1, a2, A3, A4, A5, A6 or a7 is optionally substituted with another D-amino acid or an L-amino acid.

The peptide of the preceding item, wherein one or two of a1, a2, A3, a4, a5, a6, or a7 is replaced with a corresponding L-amino acid.

The peptide according to any preceding claim, wherein any one of a1, a2, A3, a4, a5, a6 or a7 is replaced by another D-amino acid or an L-amino acid.

The peptide of the preceding item, wherein one of a1, a2, A3, a4, a5, a6, or a7 is replaced by a corresponding L-amino acid.

The peptide according to any preceding claim, wherein a1 is t.

The peptide according to any preceding claim, wherein a1-a2 is tk or tw.

The peptide according to any preceding claim, wherein a2-A3 is ks or wf.

The peptide of any preceding item, wherein a1-a2-A3 is tks or twf.

The peptide of any preceding claim, wherein A3-a4 is sr or ft.

The peptide of any preceding item, wherein a2-A3-a4 is ksr or wft.

The peptide according to any preceding claim, wherein a4 is r or t.

The peptide according to any preceding claim, wherein a5 is f, h, i, n or w.

The peptide according to any preceding claim, wherein a5 is h, i, n or w.

The peptide according to any preceding claim, wherein a5 is i or w.

The peptide according to any preceding claim, wherein a5 is f.

The peptide according to any preceding claim, wherein a1-a2 is ww.

The peptide according to any preceding claim, wherein a6-a7 is ia.

The peptide of any preceding claim, wherein a4 is r or t and a6 is i.

The peptide of any preceding claim, wherein a6-a7 is in.

The peptide according to any preceding claim, wherein a7 is n.

A peptide according to any preceding claim, comprising tksrfX/xn, wherein X/X is i, l or y.

A peptide according to any preceding claim comprising twfX/xfin wherein X/X is i, l, m, r, s, t, v or w.

A peptide according to any preceding claim comprising twX/xrfin wherein X/X is a, c, e, f, G, h, i, k, l, m, n, p, q, r, s, t, v, w or y.

A peptide according to any preceding claim comprising tX/xsrfin, wherein X/X is p, r, s or w.

The peptide of any preceding claim, comprising X/xwftfin, wherein X/X is m or w.

A peptide according to any preceding claim, comprising wwftX/xia, wherein X/X is h, i, n or w.

A peptide according to any preceding claim, comprising wwftX/xia, wherein X/X is i or w.

A peptide according to any preceding claim, comprising a sequence selected from: tksrfX/xn, tX/xsrfin, twX/xrfin, twfX/xfin, X/xwftfin, wwftfiX/X, wwftX/xia and tksrfdn or derivatives thereof, wherein X/X is glycine, or any D-amino acid, or any L-amino acid.

A peptide according to any preceding claim, comprising a sequence selected from: twftfin, X/xwftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftX/xn, twftfiX/X, and X/xftfix/X, wherein X/X is glycine, or any D-amino acid, or any L-amino acid.

A peptide according to any preceding claim, comprising a sequence selected from: twftfin, X/xwftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftfX/xn, twftfiX/X and X/xftfix/X, wherein X/X is glycine, or any D-amino acid, or any L-amino acid, and wherein, in each sequence, one other amino acid is inserted, deleted or changed to a different D-amino acid, glycine, or any L-amino acid, by one amino acid.

A peptide according to any preceding claim, comprising a sequence selected from: PSP IV A17, PSP V A19, and PSPVI A1.

A peptide according to any preceding claim comprising the sequence tksrfX/xn (formula I).

A peptide according to any preceding claim comprising the sequence tX/xsrfin (formula II).

A peptide according to any preceding claim comprising the sequence twX/xrfin (formula V).

A peptide according to any preceding item comprising the sequence twfX/xfin (formula VI).

The peptide according to any preceding claim, comprising the sequence X/xwftfin (formula VII).

A peptide as claimed in any preceding item comprising the sequence wwftfiX/x (formula VIII).

A peptide as claimed in any preceding item which comprises the sequence wwftX/xia (formula IX).

The peptide of any preceding claim, comprising the sequence X/xwftfiX/X (formula X).

The peptide of any one of the preceding claims, further comprising an N-terminal and/or C-terminal cysteine residue.

The peptide of any preceding claim which is cyclic.

The peptide of any one of the preceding claims, further comprising one or more solubility tags, each solubility tag being independently selected.

The peptide according to any preceding claim, having a solubility tag at the N-and/or C-terminus.

The peptide according to any preceding item, wherein the solubility tag is selected from the group consisting of RRKK and RRKK.

The peptide of any one of the preceding or following items, wherein the peptide has antiviral activity against retrovirus, lentivirus, paramyxovirus, flavivirus, hepatitis c virus, tick-borne encephalitis, yellow fever, dengue virus, filovirus, togavirus, bunyavirus, herpesvirus, hepnavirus, paramyxovirus, or coronavirus.

The peptide of any one of the preceding or following items, wherein the peptide has antiviral activity against herpes simplex virus, cytomegalovirus, respiratory syncytial virus, or human immunodeficiency virus.

The peptide of any one of the preceding or following items, wherein the peptide has antiviral activity against HIV, ebola virus, marburg virus, rubella virus, hantavirus, arenavirus, cytomegalovirus, hepatitis b virus, or circovirus.

The peptide of any one of the preceding or following items, wherein the infection is AIDS, herpes or SARS.

The peptide according to any one of the preceding or following items, wherein the peptide has antiviral activity against herpes simplex virus, cytomegalovirus, respiratory syncytial virus and/or human immunodeficiency virus.

The peptide of any one of the preceding or following items, wherein it is directed against the IC of a virus50Less than 100. mu.M.

The peptide according to any preceding claim, wherein X/X is selected from any proteinogenic amino acid.

A peptide according to any preceding claim wherein X/X is selected from the D-isomers of any proteinogenic amino acid.

A peptide according to any preceding item selected from ctksrfX/xnc, rrkkctX/xsrfinc, RRKKctwX/xrfin, RRKKctwfX/xffin, RRKKcX/xwffin, RRKKcwftfix/xc, RRKKcwftX/xac and ctksrfdn c.

The peptide according to any preceding claim, wherein x is selected from a, c, d, e, f, h, i, k, l, m, n, p, q, r, s, t, v, w, y or G.

The peptide according to any of the preceding items, wherein the peptide comprises the sequence RRKKctwX/xrfin, or is selected from the group consisting of RRKKcttwwarfin, RRKKctwcrcfin, RRKKctwdrfin, RRKKctwerfin, RRKKctwfrfin, RRKKctwGrfin, RRKKctwhrfin, RRKKctwrfin, RRKKctwwrfin, RRKKctwmrfin, RRKKctwrrfin, RRKKctwwrfin, RRKKctwqrfin, RRKKctwrrfin, RRKKctwrfin, RRKKctwwrfin, or RRKKctwtyrfin.

The peptide according to any of the preceding items, wherein the peptide comprises the sequence RRKKctwffX/xfinc or is selected from the group consisting of RRKKctwfaffacinc, RRKKctwffcinc, RRKKctwfdfinc, RRKKctwfeffect, RRKKctwfgfnfinc, RRKKctwfgffacmc, RRKKctwfhfinc, RRKKctwfifinc, RRKKctwfkfkfinc, RRKKctwfflfac, RRKKctwfmfinc, RRKKctwfmfnfinc, RRKKctwfnfinc, RRKKctwfffinc, RRKKctwffwfcnfinc, RRKKctwffwinc, and RRKKctwfyfyfinc.

A peptide according to any preceding claim, wherein the peptide comprises the sequence RRKKcX/xwftfinc, or is selected from RRKKcawftfinc, RRKKccwfftfinfet, RRKKcdwftfinc, RRKKcewfftfinfet, RRKKcfwftfinc, RRKKcGcgwftfc, RRKKchwfftfinfet, RRKKciwftfc, RRKKckwftfc, RRKKclwfftfinfet, RRKKcmwftfc, RRKKcnwftfc, RRKKcpwftfc, RRKKcrwftfc, RRKKcswftfc, RRKKctwftfc, RRKKcvftfinfet, RRKKcckwftfinfet, RRKKcvftfinfet, and RRKKcwftfinfet.

A peptide according to any one of the preceding items, wherein the peptide comprises the sequence RRKKcwftfix/xc or is selected from RRKKcwftfix, RRKKcwwftffic, RRKKcwwftfidc, RRKKcwwftffic, RRKKcwftfic, RRKKcwwftffic, RRKKcwftfic, RRKKcwftficc, RRKKcwftfic, RRKKcwftvcftvcftvcftvcftfic, RRKKcwwKKwKKwKKftfic, RRKKwKKwKKwKKwKKftfic, RRkwftfic, RRKKcwwKKwffic, RRkwftfic, RRKKcwwffic, and RRKKcwwfiffic.

The peptide according to any of the preceding items, wherein the peptide is selected from ctksrfanc, ctksrfcnc, ctksrfdnc, ctksrffennc, ctksrffnc, ctksrfGnc, ctksrfhnc, ctksrfinc, ctksrfknc, ctksrflnc, ctksrfmnc, ctksrffnc, ctksrfqnc, ctksrffnnc, ctksrfvnc, ctksrfcnc, ctksrfntnc, ctksrfvnc and ctksrfync.

The peptide of any of the preceding items, wherein the peptide is selected from the group consisting of rrkkctasrfinc, rrkkctcscrfinc, rrkkctdsrfinc, rrkkctesrfinc, rrkkctfsrfinc, rrkkctGsrfinc, rrkkcthsrfinc, rrkkctisrfinc, rrkkctksrfinc, rrkkctlsrfinc, rrkkctmsrfinc, rrkkctnsrfinc, rrkkctpsrfinc, rrkkctqsfinc, rrkkctrsfinc, rrkkctssrfinc, rrkkcttsfinc, rrkkctvsfinc, rrkkctwsfinc, and rrkkctysfinc.

A peptide according to any preceding claim, wherein the peptide is selected from RRKKcwftaiac, RRKKcwftciac, RRKKcwftdiac, RRKKcwwfteiac, RRKKcwftgiaac, RRKKcwftkiac, RRKKcwftliac, RRKKcwftmiac, RRKKcwftniac, RRKKcwftpiac, RRKKcwftgiac, RRKKcwftriac, RRKKcwftsiac, RRKKcwftgiac and RRKKcwftgiac.

The peptide of any preceding claim, wherein the peptide comprises twfrfin, twfffin, twftfin, wwftfin and wwftfia.

The peptide of any preceding claim, wherein the peptide comprises ctwfrfinc, ctwfffpic, ctwfftfinfet, cwftfinfet and cwftfiac.

A peptide according to any preceding claim wherein the peptide is selected from RRKKctwfrfinc, RRKKctwfffpic, RRKKctwftfinc, RRKKcwftfinc and RRKKcwwftfiac.

A peptide according to any preceding claim for use in the preparation of a medicament.

A peptide according to any preceding claim for use as a medicament.

A pharmaceutical composition comprising a peptide according to any preceding claim.

A peptide according to any preceding claim, or a pharmaceutical composition thereof, suitable for use in the treatment of a viral infection.

Use of a peptide according to any preceding claim, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a viral infection.

A method of treating a viral infection in a host animal, comprising administering to the host animal a therapeutically effective amount of a peptide according to any one of the preceding claims, or a pharmaceutical composition thereof.

The method of any of the preceding items, wherein the peptide or pharmaceutical composition is administered by nasal, pulmonary, intrabronchial, oral or parenteral route.

The method of any of the preceding claims, wherein the peptide or pharmaceutical composition is administered prophylactically.

A method for detecting a peptide having an antiviral activity, which comprises preparing a derivative of the peptide of any one of the preceding items and screening for an antiviral activity.

A kit comprising a peptide according to any preceding claim, or a pharmaceutical composition thereof, and instructions for use of the peptide or pharmaceutical composition in the treatment of a viral infection.

A vector comprising a nucleic acid encoding a peptide according to any preceding claim.

A plasmid comprising a nucleic acid encoding a peptide according to any preceding claim.

A host cell expressing a peptide according to any preceding claim.

A combination comprising a peptide of any one of the preceding items and a second agent selected from the group consisting of a nucleic acid, a peptide, a protein, a contrast agent, an antibody, a toxin, and a small molecule.

A recombinant library comprising a peptide according to any preceding claim.

Without being bound by theory, it is also believed herein that the peptides disclosed herein comprise membrane switch motifs that disrupt the viral lipid envelope in order to achieve access to underlying matrix proteins, which may play a key role in their antiviral activity. Thus, the peptides of the invention are expected to bind to and inhibit any lipid envelope-containing virus, including but not limited to: togaviridae (Togaviridae), including rubella virus; the family retroviridae, including lentiviruses such as HIV; bunyaviridae (Bunyaviridae), such as hantavirus and arenavirus; herpesviridae, such as herpes virus and cytomegalovirus; hepnaviridae, such as hepatitis b virus; paramyxoviridae, such as paramyxovirus; flaviviridae (Flaviviridae), such as flavivirus, hepatitis c virus, tick-borne encephalitis, yellow fever, and dengue virus; filoviridae (Filoviridae), such as ebola and marburg viruses; coronaviridae (Coronaviridae), such as coronaviruses, including SARS virus and circovirus.

Derivatives of twftfin include peptides in which one or more amino acids are deleted or substituted with a different amino acid, as shown in table 1. Derivatives also include peptides having at least 80%, 85%, 90% or 95% sequence identity. The derivative preferably comprises, or consists essentially of, 7 amino acids. In another embodiment, the derivative comprises a7 amino acid core peptide, and further comprises additional amino acids to improve various properties, solubility, or achieve cyclization as described herein. The core peptide is an active peptide, i.e. a peptide that confers antiviral activity. The derivatives are functional, that is, they are biologically active and preferably exhibit antiviral properties against more than one virus.

As used herein, the term "peptide" refers to a polymer comprising amino acid residues. In exemplary embodiments, the peptide comprises from 2 to about 100 residues or from 2 to about 50 residues. In other embodiments, the peptide comprises about 7 to 20 residues, e.g., 7 to 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19 residues. In certain embodiments, the amino acid residues comprising the peptide are all "D-type" amino acid residues, however, it is recognized that in various embodiments, "L" amino acids may be included in the peptide. Thus, in some embodiments, the peptides of the invention are a mixture of D-and L-form amino acids. For example, the peptide may comprise a core of 7D-amino acids, one or more of which may be in the L form.

In addition, the term applies to amino acids linked by peptide bonds or other "modified bonds" (e.g., peptide bonds replaced by a-esters, β -esters, thioamides, phosphonamides, carbamates, hydroxylates, and the like).

In certain embodiments, the peptides of the invention comprise an active core peptide having 7 residues. In addition, the peptide may comprise additional residues, including but not limited to one or more residues that may allow the peptide to form a cyclic peptide and/or one or more residues that may increase the solubility of the peptide as described herein.

The peptides described herein may be naturally occurring or non-naturally occurring. It can be produced a) by chemical synthesis, b) by recombinant DNA techniques, c) by biochemical or enzymatic cleavage of larger molecules, d) by a combination of any of the methods listed above from a to d, e) by any other method of producing peptides. In another embodiment, the peptide is non-naturally occurring. In another embodiment, the peptide comprises a non-naturally occurring modification. Examples of such modifications are described herein.

Table 1: an exemplary family of twftfin derivatives; for clarity, the N and C terminals are shown.

Figure BDA0002336262210000191

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of tksrfX/xn, tX/xsrfin, twX/xrfin, twfX/xfin, X/xwftfin, wwftfiX/X, wwftX/xia, and tksrfdn. In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of tksrfX/xn, tX/xsrfin, twX/xrfin, twfX/xfin, X/xwftfin, wwftfiX/X, and wwftX/xia.

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of twftfin, X/xftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftfX/xn, twftfX/X, and X/xwftfiX/X, wherein X/X is glycine, or any D-amino acid, or any L-amino acid.

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of twftfin, X/xftfin, tX/xftfin, twX/xtfin, twfX/xfin, twftX/xin, twftfX/xn, twftfiX/X and X/xftfix/X, wherein X/X is glycine, or any D-amino acid, or any L-amino acid, and wherein one additional amino acid is inserted, deleted or changed in each sequence to a different D-amino acid, glycine or any L-amino acid.

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from ctksrfX/xnc, ctX/xsrfinc, ctwX/xrfinc, ctwfX/xfinc, cX/xwftfinc, cwftfix/xc, cwftx/xiac, and ctksrfdnc. In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from ctksrfX/xnc, ctX/xsrfinc, ctwX/xrfinc, ctwfX/xfinc, cX/xwftfinc, cwftfix/xc, and cwftx/xiac.

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of RRKKctksrfX/xnc, RRKKctX/xsfinc, RRKKctwX/xrfinc, RRKKctwfX/xffinc, RRKKcX/xwffinc, RRKKcwwftfiX/xc, rrkkcwftx/xiac, and RRKKctksrfdnc. In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of RRKKctksrfX/xnc, RRKKctX/xsfinc, RRKKctwX/xrfinc, RRKKctwfX/xfinc, RRKKcX/xwftfinc, rrkkcwftfix/xc, and rrkkcwftx/xiac.

In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of rrkkctksrfX/xnc, rrkkctX/xsfinc, rrkkctwX/xrfinc, rrkkctwfX/xfinc, rrkkcX/xwffinc, rrkkcwwftfiX/xc, rrkkcwftx/xiac, and rrkkctksrfdnc. In another embodiment, an antiviral peptide is described comprising, consisting essentially of, or consisting of a sequence selected from the group consisting of rrkkctksrfX/xnc, rrkkctX/xsrfin, rrkkctwX/xrfin, rrkkctwfX/xfinc, rrkkcX/xwfflnc, rrkkcwwftfiX/xc, and rrkkcwftx/xiac.

It is to be understood that in any of the preceding embodiments, the D-cysteine may be L-cysteine.

In the sequences described herein, the amino acid name "X/X" indicates the position at which a substitution is made, which may include any L-amino acid or D-amino acid or any amino acid analog. The letter "X/X" denotes any proteinogenic amino acid, i.e. any of the 20 naturally occurring amino acids. Except for achiral G, X represents the amino acid in L form and X represents the amino acid in D form. In another embodiment, X/X is X, i.e., the amino acid in D form, or X, i.e., the amino acid in L form. In another embodiment, x is selected from the following amino acids in D form: a. c, d, e, f, h, i, k, l, m, n, p, q, r, s, t, v, w or y. In another embodiment, X is selected from the following amino acids in the form of L: A. c, D, E, F, H, I, K, L, M, N, P, Q, R, S, T, V, W or Y. In another embodiment, X is glycine.

In another embodiment, the IC of the peptide50Less than 100. mu.M, or less than 50. mu.M, or less than 10. mu.M, or less than 5. mu.M, or less than 1. mu.M, or less than 0.1. mu.M. In another embodiment, the peptide exhibits an inhibition of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more of at least one virus and/or at least one virus subtype, preferably of at least two viruses and/or at least two virus subtypes. Preferably, the inhibition is at least 50%, 60%, 70%, 80%, 90%, 95% or more, or at least 75%80%, 90%, 95% or higher.

In another embodiment, the peptide has antiviral activity against preferably 2, 3, 4 or more viruses and/or virus subtypes, e.g., as measured in a plaque reduction assay on MDCK cells using, e.g., 0.01 μ M to 100 μ M peptide. In another embodiment, the peptide exhibits at least 75%, 80%, 90% or 95% inhibition of at least two viral subtypes.

In another embodiment, the peptides of the invention comprise one or more solubility tags. In an exemplary embodiment, the solubility tag is a short peptide consisting of charged amino acids that, when covalently linked to the N-terminus or C-terminus of a hydrophobic peptide, will increase the solubility of the linked peptide. For example, the solubility tag may comprise one or more amino acids selected from D, E, H, K, N, Q, R, S, T, hydroxyproline, and pyroglutamic acid because of the hydrophilic nature of these amino acids. These amino acids may be in the levorotatory or dextrorotatory form. The addition of any of these amino acids, particularly E/D/R/K, in any combination or repetition at the N-or C-terminus can be used to increase the solubility of the peptide. Solubility may also be increased by the addition of dipeptide EE, tripeptide SGS, and/or by hyperglycosylation and pegylation or other methods known in the art to increase solubility.

For example, the solubility tag may comprise or consist of a solubility tag RRKK (in levorotatory form RRKK or dextrorotatory form RRKK) with the N-terminus and/or C-terminus of the peptide.

The peptides of the invention may be in linear or cyclic form. In another embodiment, the peptide of the invention is in a cyclic form. The peptide may be cyclized, for example, by forming a disulfide bond between cysteine residues (or more generally, between two of at least two cysteine residues present in the polypeptide (e.g., in a terminal region)). It will be appreciated that each cysteine residue is independently D-cysteine or L-cysteine. For example, at least two cysteine residues may be added, one or both of which are optionally at the C-terminus or N-terminus of the peptide. Thus, antiviral peptides and derivatives thereof may be cyclized with their terminal cysteine residues via a disulfide bond. As shown herein, the linear form of the peptide (i.e., without any cyclization) exhibits antiviral properties. It will also be appreciated that where the peptide contains three or more cysteine residues, one or more of the residues may be modified to prevent the residue from participating in the formation of a disulphide bond. This may conveniently be achieved by protecting the thiol group of cysteine with an acetamidomethyl group to form the cysteine analogue acetamidomethyl-cysteine (cys (acm)). In the context of the peptides of the invention, this can be done with internal cysteine residues. The modification may be performed during synthesis (i.e., the modified residue is used for synthesis), or may be performed post-synthesis.

Strategies for preparing cyclic polypeptides from linear precursors have been described and may be used with the peptides of the invention. For example, methods include chemical cross-linking, chemical intra-molecular attachment methods, and enzymatic intra-molecular attachment methods that allow linear synthetic peptides to be efficiently cyclized under aqueous conditions.

In another embodiment, described herein are peptides that are in cyclic form and comprise a solubility tag, such as RRKK (levorotatory or dextrorotatory form).

In another embodiment, the peptide is selected from ctksrfX/xnc, rrkkctX/xsrfinc, RRKKctwX/xrfin, RRKKctwfX/xffin, RRKKcX/wftfinc or RRKKcwftfix/xc, RRKKcwftX/xac, or ctksrfdnc.

In another embodiment, the peptide of the invention comprises the general sequence twX/xrfin (formula V), wherein X/X is a, c, d, e, f, l, m, n, p, q, r, s, t, V, w, or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of rrkkctwarfin, RRKKctwcrfinc, RRKKctwfrfinc, RRKKctwrrfinc, and RRKKctwrrfinc. Peptides as described above with the solubility tag in the D form (rrkk) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 2: exemplary peptides

Figure BDA0002336262210000231

In another embodiment, the peptides of the invention comprise the universal sequence twfX/xfin (formula VI), wherein X/X is a, c, d, e, f, G, h, i, k, l, m, n, p, q, r, s, t, v, w, or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of RRKKctwfafinc, rrkkctwffcinc, RRKKctwfdfinc, RRKKctwfefinc, RRKKctwfffinc, RRKKctwfGfinc, RRKKctwfhfinc, rrkkctwfefinic, rrkkctwfkfkfkfinc, RRKKctwfnfinc, rrkkctwffpinc, RRKKctwfffinc, and rrkkctwfyfnfinc. Peptides as described above with the solubility tag in the D form (rrkk) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 3: exemplary peptides

Figure BDA0002336262210000251

In another embodiment, the peptide of the invention comprises the general sequence X/xwftfin (formula VII), wherein X/X is a, c, d, e, f, G, h, i, k, l, m, n, p, q, r, s, t, v, w or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of RRKKcawftfinc, RRKKccwftfpic, RRKKcdwftfpic, RRKKcewftfpic, RRKKcfwftfpic, RRKKcGwftfpic, RRKKchwftfpic, RRKKciwftftftfpic, RRKKckwftftftfpic, RRKKclwftftfpic, RRKKcmwftfpic, RRKKcnwftfpic, RRKKcpwftfpic, RRKKcqwfpic, RRKKcrwftfpic, RRKKcswftfpic, RRKKctwftfpic, RRKKcvftpic, RRKKcwwftfpic, RRKKcwftpic, and RRKKcwftpic. Peptides as described above with the solubility tag in the D form (rrkk) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 4: exemplary peptides

Figure BDA0002336262210000252

Figure BDA0002336262210000261

In another embodiment, the peptide of the invention comprises the general sequence wwftfiX/X (formula VIII), wherein X/X is a, c, d, e, f, G, h, i, k, l, m, p, q, r, s, t, v, w or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of RRKKcwftfiac, RRKKcwftficc, RRKKcwftfidc, RRKKcwwftfficc, RRKKcwwftfiftfficc, RRKKcwftfidc, RRKKcwftfilc, RRKKcwftfimc, RRKKcwftfipc, RRKKcwftfiqc, RRKKcwftfidc. Peptides as described above with the solubility tag in the D form (rrkk) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 5: exemplary peptides

Figure BDA0002336262210000262

Figure BDA0002336262210000271

In another embodiment, the peptides of the invention comprise the general sequence tksrfX/xn (formula I), wherein X/X is a, c, d, e, f, G, h, I, k, l, m, n, p, q, r, s, t, v, w or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from ctksrfanc, ctksrfcnc, ctksrfdnnc, ctksrffunc, ctksrffnc, ctksrfnc, ctksrfnfnc, ctksrfnknc, ctksrflnc, ctksrfnfmnc, ctksrfqnc, ctksrfrnc, ctksrfrnnc, ctksrfvnc, ctksrfncc, ctksrfnvnc, ctksrfnc, and ctksrfync. Peptides as described above having a solubility tag in the L form (RRKK) or the D form (RRKK) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 6: exemplary peptides

Figure BDA0002336262210000272

In another embodiment, the peptides of the invention comprise the general sequence tX/xsrfin (formula II), wherein X/X is a, c, d, e, f, G, h, i, k, l, m, n, p, q, r, s, t, v, w or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of rrkkctasrfinc, rrkkctcsrfinc, rrkkctdsrfinc, rrkkctesrfinc, rrkkctfsrfinc, rrkkctGsrfinc, rrkkcthsrfinc, rrkkctssrfinc, rrkkctksrfinc, rrkkctlsrfinc, rrkkctmsrfinc, rrkkctnsfinc, rrkkctpsrfinc, rrkkctqsfinc, rrkkctrsfinc, rrkkctssrfinc, rrkkcttsrfinc, rrkkcttsfinc, and rrkkctysfinc. Peptides as described above having the solubility tag in the L form (RRKK) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 7: exemplary peptides

Figure BDA0002336262210000291

In another embodiment, the peptide of the invention comprises the general sequence wwftX/xia (formula IX), wherein X/X is a, c, d, e, G, h, i, k, l, m, n, p, q, r, s, v, w or y. The peptides may comprise cysteine residues at the N and C termini for cyclization and/or solubility tags. In another embodiment, the peptide is selected from the group consisting of RRKKcwftaiac, RRKKcwftciac, RRKKcwftdiac, RRKKcwfteiac, RRKKcwwfftgiaac, RRKKcwfthiaac, RRKKcwftiac, RRKKcwftkiac, RRKKcwftliac, RRKKcwftmiac, RRKKcwftniac, RRKKcwftpiac, RRKKcwftgiac, RRKKcwftriac, RRKKcwftsiac, RRKKcwftgiac, RRKKcwftwiac, and RRKKcwftyiac. Peptides as described above with the solubility tag in the D form (rrkk) are also within the scope of the invention. Functional derivatives of these peptides, without any cyclisation, are also within the scope of the invention.

Table 8: exemplary peptides

Figure BDA0002336262210000301

In another embodiment, the peptides described herein have antiviral activity against at least one, preferably more than one, virus that infects humans. In another embodiment, the peptides described herein have antiviral activity against more than one viral subtype (the subtype that preferentially infects humans). For example, the peptides described herein can be used to treat infections caused by HIV, RSV, various subtypes of HSV and CMV including, but not limited to, RSV A2, HSV-2, CMV AD 169, HIV-192 UG029, and the like. In another embodiment, the virus is not an influenza virus or an orthomyxovirus.

The signature peptides and biologically active variants thereof can be modified in a variety of ways. For example, reagents including additional amino acid residues, other substituents, and protecting groups can be added to the amino terminus, the carboxyl terminus, or both. For example, as described above, the peptides may be modified to include cysteine residues or other sulfur-containing residues or agents that may be involved in disulfide bond formation.

The peptides of the invention may also comprise one or more modified amino acids at any position(s) and/or N-and/or C-terminus to increase solubility, stability, reactivity or improve other properties. These modifications include the addition of chemical groups such as, but not limited to, acetyl, benzyloxycarbonyl, dansyl, tert-butoxycarbonyl, 9-fluorenylmethoxycarbonyl or hydrophilic groups to the amino terminus and/or to the tert-butoxycarbonyl group, or the addition of amide groups or p-nitrobenzyl ester groups or hydrophilic groups to the carboxyl terminus. They may also contain modified amide linkages such as N-methylation.

In another embodiment, any of the peptides described herein comprises one or more substituents. For example, the peptide may comprise substituents at the amino terminus, the carboxyl terminus, and/or on the side chains of reactive amino acid residues. The substituent may be an acyl group or a substituted or unsubstituted amine group (for example, the substituent at the N-terminus may be an acyl group, and the C-terminus may be amidated with a substituted or unsubstituted amine group (for example, an amino group having one, two, or three substituents which may be the same or different)). The amine group can be a lower alkyl (e.g., an alkyl having 1-4 carbons), alkenyl, alkynyl, or haloalkyl. The acyl group may be a lower acyl group (e.g., an acyl group having up to four carbon atoms), particularly an acetyl group. The substituents may be non-protein polymers such as polyethers, polyethylene glycol (PEG), polypropylene glycol or polyalkylene oxide, polyalkylene glycols (e.g. polypropylene glycol (PPG), polybutylene glycol (PBG), or PPG-PEG block/random polymers).

The size and shape of the non-protein polymer may vary. For example, any of the non-protein polymers listed above (e.g., PEG) may be linear, branched, or comb-shaped. With respect to size, the molecular weight may vary. For example, the molecular weight of the PEG can be, for example, about 300Da, about 1000Da, about 2000Da, about 3000Da, about 4000Da, about 5000Da, about 6000Da, about 7000Da, about 8000Da, about 9000Da, about 10000Da, about 11000Da, about 12000Da, about 13000Da, about 14000Da, about 15000Da, about 20000Da, about 30000Da, about 40000Da, or about 50000 Da. For example, the molecular weight of the PEG can be any value between 300Da and 2000Da, between 300Da and 3000Da, between 1000Da and 2000Da, and between 1000 and 3000 Da. Non-protein polymers (e.g., PEG) can be attached to the peptide by any number of functional group chemicals (e.g., carboxylated mPEG, p-nitrophenyl-PEG, aldehyde-PEG, amino-PEG, thiol-PEG, maleimide-PEG, aminoxy-PEG, hydrazine-PEG, tosyl-PEG, iodoacetamide-PEG, succinimidyl succinate-PEG, succinimidyl glutarate-PEG, succinimidyl carboxypentyl-PEG, p-nitrophenyl carbonate-PEG, or ethanethiol-PEG). Non-protein polymers (e.g., PEG) can be attached to the peptide through any number of chemical groups including, but not limited to, amino-terminal amino acids, carboxy-terminal amino acids, free amines, and free thiols.

Non-protein polymers (e.g., PEG) can be functionalized (e.g., monofunctional activated linear PEG, homobifunctional activated linear PEG, heterobifunctional activated linear PEG, multi-arm activated PEG (e.g., 2-arm, 4-arm, 8-arm, etc.), branched activated PEG, and comb activated PEG).

The antiviral peptide of the present invention may further comprise a reactive tag at the terminal amino acid thereof for the purpose of detection, isolation, purification, etc. These tags may include, but are not limited to, biotin, histidine, GST, and the like. Suitable labels are known in the art.

The peptides of the invention may also be linked to lipids (phospholipids) or polyethylene glycols to enhance/modify solubility, proteolytic stability or other properties and/or activity. Accordingly, the peptides of the present invention include such lipopeptides.

As used herein, the antiviral peptides of the present invention may include mimetics or peptidomimetic derivatives, which are compounds capable of mimicking the core structure of a peptide. Similarly, it will be appreciated that the antiviral peptide may include one or more amino acid substitutions, such as isosteres or bioisosteres.

In another embodiment, the antiviral peptides of the present invention may be in multimeric form. The multimer may be a dimer, trimer or tetramer; preferably a dimer. The monomeric antiviral peptides forming the multimer may be composed of the same or different peptides. In a preferred embodiment, the peptides forming the multimer are covalently linked.

In another embodiment, the antiviral peptides of the invention may exist in different physical forms. The physical form may be crystalline, amorphous, or other forms well known in the art.

The various examples provided in this patent application demonstrate that FPT with substituted amino acids retains antiviral activity as shown by the various sequences described in tables 1-8. Such derivatives are within the scope of the present invention. In another embodiment, amino acids of the antiviral peptides of the present invention may be substituted by conservative substitutions. Conservative substitutions are those that replace an amino acid with a substitute amino acid that changes the primary sequence but does not alter function. Examples of conservative amino acid substitutions include: valine, isoleucine and leucine; lysine and arginine; asparagine and glutamine; serine and threonine; glycine and alanine; phenylalanine and tyrosine, and aspartic acid and glutamic acid. Amino acids within a given group may be conservatively substituted with another amino acid in the same group.

In another embodiment, the amino acids of the antiviral peptides of the present invention may be substituted with non-natural or unnatural (non-natural or unnatural) amino acids and derivatives thereof, including, but not limited to, β -amino acids (β 3 and β 2), Homo-amino acids (Homo-amino acids), proline and pyruvate derivatives, 3-substituted alanine derivatives, glycine derivatives, ring-substituted phenylalanine and tyrosine derivatives, N-methyl amino acids, and other amino acid derivatives well known in the art.

The peptides of the present invention or derivatives thereof can be tested for antiviral activity using the methods explained in the examples below.

The disclosed peptides and derivatives thereof can be synthesized by any of the usual methods used in peptide synthesis, such as liquid or solid phase synthesis, or any other method well known in the art.

The peptides of the present invention exhibit a broad spectrum of antiviral activity against viruses including various types and/or subtypes. The virus may be of human or animal origin, for example of simian origin. Preferred viruses are of human origin or human infectious viruses.

Thus, according to another aspect of the invention, one or more peptides of the invention as described above may be formulated into a pharmaceutical composition. The composition may comprise at least one peptide of the invention and one or more pharmaceutically acceptable carriers, including excipients, such as diluents, carriers, and the like, and additives, such as stabilizers, preservatives, solubilizers, buffers, and the like, as desired. A pharmaceutically acceptable carrier is a compound that can be administered with the peptides of the invention and that contains one or more non-toxic ingredients that do not inhibit or reduce the effectiveness of the active antiviral peptides described herein.

The pharmaceutically acceptable carrier or excipient may be particulate such that the composition is in the form of, for example, a tablet or powder. The carrier may be a liquid and the composition may be, for example, an oral syrup or an injectable liquid. Alternatively, the carrier may be gaseous so as to provide an aerosol composition which may be used, for example, for administration by inhalation.

Such pharmaceutical carriers can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The carrier can be saline, gum arabic, gelatin, starch paste, talc, keratin, colloidal silica, urea, etc. In addition, adjuvants, stabilizers, thickeners, lubricants and colorants may be used. In another embodiment, the peptide or composition of the invention and the pharmaceutically acceptable carrier are sterile when administered to an animal. Water is a preferred carrier when the peptides of the invention are administered intravenously. Saline solutions, as well as aqueous dextrose and glycerol solutions, may also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The compositions of the present invention may also contain minor amounts of wetting or emulsifying agents or pH buffering agents, if desired.

In another embodiment, specific non-limiting examples of carriers and excipients include, but are not limited to, water, saline, dextrose, glycerol, ethanol, and the like, or combinations thereof. Formulation excipients may include polyvinylpyrrolidone, gelatin, hydroxycellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.

As a solid composition for oral administration, the composition may be formulated into the form of powder, granules, compressed tablets, pills, capsules, chewing gum, flakes, and the like. Such solid compositions typically comprise one or more inert diluents. Furthermore, there may be one or more of the following: binders, such as carboxymethyl cellulose, ethyl cellulose, microcrystalline cellulose or gelatin; excipients, such as starch, lactose or dextrin, disintegrants, such as alginic acid, sodium alginate, corn starch, and the like; lubricants, such as magnesium stearate; glidants such as colloidal silicon dioxide; sweetening agents, such as sucrose or saccharin; flavoring agents, such as peppermint, methyl salicylate, or orange flavoring; and a colorant.

For topical formulations, any of a variety of creams, ointments, gels, lotions and the like may be used.

When used for oral administration, the compositions are preferably in solid or liquid form, wherein semi-solid, semi-liquid, suspension and gel forms are included as solid or liquid in the forms contemplated herein.

The compositions may be in the form of a liquid, such as an elixir, syrup, solution, emulsion or suspension. The liquid can be used for oral administration or delivery by injection. When used for oral administration, the compositions may contain one or more of sweetening agents, preserving agents, dyes/colorants and taste-enhancing agents. In the composition for injection administration, one or more of a surfactant, a preservative, a wetting agent, a dispersing agent, a suspending agent, a buffer, a stabilizer, and an isotonic agent may also be included. Water containing at least one or more buffering ingredients is preferred, with or without stabilizers, preservatives and solubilizers. When the composition is in the form of a capsule (e.g. a gelatin capsule), it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol, cyclodextrin or a fatty oil.

For most pharmaceutical formulations, the inactive ingredients will make up a larger portion of the weight or volume of the formulation. For pharmaceutical formulations, it is also contemplated that any of a variety of measured release, slow release, or time release formulations may be used, as well as additives, such that a dosage form may be formulated to achieve effective delivery of the peptides of the invention over a period of time.

It will be appreciated that the peptides described herein may be administered using any suitable route of administration or method of delivery, including but not limited to oral, parenteral, depot, buccal, and the like.

In certain embodiments, the peptide(s) are formulated with a non-covalent carrier. Non-covalent carriers are well known to those skilled in the art.

In certain embodiments, the peptides are complexed with carriers such as lipids or formulated into liposomes, biodegradable or non-biodegradable microparticles, microcapsules, microspheres, and nanoparticles. Methods for producing liposomes, microparticles, microcapsules, microspheres, and nanoparticles, and for complexing or encapsulating compounds therein, are well known to those skilled in the art.

In certain embodiments, the antiviral peptides described herein are formulated as a nanoemulsion. Nanoemulsions include, but are not limited to, oil-in-water (O/W) nanoemulsions and water-in-oil (W/O) nanoemulsions. Nanoemulsions can be defined as emulsions having an average droplet diameter of from about 20 to about 1000 nm. Typically, the average droplet size is between about 20nm or 50nm to about 500 nm.

Exemplary oil-in-water (O/W) and/or water-in-oil (W/O) nanoemulsions include, but are not limited to:

1) surfactant micelles-micelles consisting of small molecule surfactants or detergents (e.g. SDS/PBS/2-propanol) which are suitable for peptides which are predominantly hydrophobic;

2 polymeric micelles-micelles consisting of a polymer, copolymer or block copolymer surfactant (e.g. pluronic l 64/PBS/2-propanol) which are suitable for peptides which are predominantly hydrophobic;

3) mixed micelle: micelles in which more than one surfactant component is present or in which one of the liquid phases (typically an alcohol or fatty acid compound) participates in the formation of micelles (e.g. caprylic acid/PBS/EtOH) are suitable for peptides that are predominantly hydrophobic;

4) whole peptide micelles — mixed micelles, where the peptide serves as a co-surfactant, form part of a micelle (e.g., amphiphilic peptide/PBS/mineral oil), which is suitable for amphiphilic peptides; and

5) pickering (solid phase) emulsion-an emulsion in which a peptide is attached to the outside of a solid nanoparticle (e.g., polystyrene nanoparticle/PBS/oil-free phase), which is suitable for amphiphilic peptides.

In another embodiment, the invention relates to a peptide or composition of the invention for use as a medicament.

Administration of the peptides and compositions of the invention includes, but is not limited to, oral, topical, parenteral, sublingual, rectal, vaginal, ocular, pulmonary (inhalation), and intranasal administration. Parenteral administration includes subcutaneous injection, intravenous, intramuscular, intrasternal injection or infusion techniques. Preferably, the composition is administered parenterally, pulmonary or orally. The pharmaceutical compositions of the invention may be formulated such that the peptides of the invention are bioavailable upon administration of the composition to an animal, preferably a human. The composition may take the form of one or more dosage units, wherein for example a tablet may be a single dosage unit and a container containing a peptide of the invention in aerosol form may contain a plurality of dosage units.

Parenteral administration includes, but is not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, intracerebral, intraventricular, intrathecal, intravaginal, or transdermal administration. The preferred mode of administration is at the discretion of the practitioner and will depend in part on the site of the medical condition.

The liquid compositions of the present invention, whether in solution, suspension or other similar form, may also include one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline, ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono-or diglycerides, polyethylene glycols, glycerol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; and agents for regulating tonicity, such as sodium chloride or dextrose. The parenteral compositions may be enclosed in ampoules, disposable syringes or multiple dose vials made of glass, plastic or other material. Physiological saline is a preferred adjuvant.

The peptide or composition is administered in a therapeutically effective amount. The term "therapeutically effective amount" or "pharmaceutically effective amount" refers to the amount and/or dose and/or dosage regimen of one or more compounds necessary to produce the desired result, e.g., an amount sufficient to reduce or prevent viral reproduction, or an amount sufficient to reduce the severity of a viral disease or delay the progression of symptoms thereof (e.g., a therapeutically effective amount), an amount sufficient to reduce the risk of or delay the onset of a disease caused by a viral infection and/or reduce the ultimate severity thereof (e.g., a prophylactically effective amount).

The correct dosage of the compound will vary depending upon the particular formulation, mode of administration and the particular site, host and disease being treated. Other factors such as age, body weight, sex, diet, time of administration, rate of excretion, host condition, drug combination, reaction sensitivity and disease severity should also be considered. Administration may be continuous or periodic over a range of maximally tolerated doses.

Typically, the amount is at least about 0.01% of the peptide of the invention by weight of the composition. When used orally, the amount may range from about 0.1% to about 80% by weight of the composition. Preferred oral compositions may comprise from about 4% to about 50% of the peptide of the invention by weight of the composition.

Preferred compositions of the invention are prepared such that the parenteral dosage unit comprises from about 0.01% to about 2% or more by weight of a peptide of the invention.

For intravenous administration, the composition may comprise from about generally about 0.1 mg/day to about 250 mg/day, preferably between about 0.1 mg/day and about 50 mg/day.

The peptides may be administered therapeutically or prophylactically. The appropriate treatment is administered 1-4 times per day, and preferably for 1-7 days or longer. As used herein, the terms "treatment", "treating" or "treatment" refer to an effect that produces a desired effect on the symptoms or pathology of a disease or disorder, particularly an effect that can be achieved using the peptides described herein, and treatment may include, but is not limited to, even minimal alteration or improvement in one or more measurable markers of the disease or disorder being treated. Treating also refers to delaying the onset, delaying or reversing the progression, reducing the severity, or reducing or preventing the disease or disorder to which the term applies, or one or more symptoms of the disease or disorder. "treating", "treating" or "treatment" does not necessarily mean completely eradicating or curing the disease or disorder or symptoms associated therewith. In another embodiment, treating comprises ameliorating at least one symptom of the treated disease. The improvement may be partial or complete. The subject receiving the treatment is any subject in need thereof. Exemplary markers of clinical improvement will be apparent to those skilled in the art.

The peptides or compositions of the invention may be administered by any convenient route, for example by infusion or bolus absorption through the epithelial or mucocutaneous linings.

Pulmonary administration can also be employed, for example, by use of an inhaler or nebulizer, and formulation with a nebulizer, or by infusion in fluorocarbon or synthetic pulmonary surfactants. In certain embodiments, the peptides or compositions of the invention may be formulated as suppositories with conventional binders and carriers such as triglycerides.

As described above, the antiviral peptide of the present invention may be formulated by any means known in the art, including, but not limited to, tablets, capsules, caplets, suspensions, powders, lyophilized formulations, suppositories, eye drops, skin patches, orally dissolvable formulations, sprays, aerosols, and the like, according to the administration route, and may be mixed and formulated with buffers, binders, excipients, stabilizers, antioxidants, and other agents known in the art.

Administration can also be carried out with another active compound, for example another antiviral compound. More than one peptide of the invention may also be administered.

The peptides described herein may also be provided as pharmaceutically acceptable salts. "pharmaceutically acceptable salts" include salts formed with: inorganic bases (e.g., alkali metals such as sodium or potassium, alkaline earth metals such as calcium and magnesium or aluminum, and ammonia), organic bases (e.g., trimethylamine, triethylamine, pyridine, picoline, ethanolamine, diethanolamine and triethanolamine), inorganic acids (e.g., hydrochloric acid, hydroboric acid, nitric acid, sulfuric acid and phosphoric acid) or organic acids (e.g., formic acid, acetic acid, trifluoroacetic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid and p-toluenesulfonic acid). The examples cited herein are non-limiting.

The invention also relates to a kit comprising a peptide or composition of the invention, instructions for use and optionally an adjuvant.

The present invention also relates to screening methods for detecting peptides having antiviral activity comprising preparing derivatives of the isolated antiviral peptides described herein, exposing the peptides to a virus and screening for antiviral activity.

The invention also relates to isolated nucleic acid molecules, including DNA or RNA molecules, that encode the antiviral peptides of the invention. In another embodiment, the isolated nucleic acid is not naturally occurring.

The invention also relates to vectors comprising nucleic acids encoding the antiviral peptides of the invention. The vector may be an expression vector, in particular a vector for expression in eukaryotic cells. Such vectors may be, for example, viral, plasmid, cosmid, or artificial chromosome (e.g., yeast artificial chromosome) vectors.

The vectors described herein can be introduced into cells or tissues by any of a variety of methods known in the art. Such methods are described, for example, in Sambrook et al, Molecular Cloning: A laboratory Manual, Cold Spring Harbor,4th edition (June 15,2012).

The invention also relates to host cells comprising the antiviral peptides of the invention.

The host cell may be any prokaryotic or eukaryotic cell, but eukaryotic cells are preferred. Exemplary eukaryotic cells include mammalian cells (e.g., Chinese Hamster Ovary (CHO) or COS cells). Other suitable host cells are known in the art.

Thus, in the present invention, cells may be transfected in vitro or ex vivo, and the expressed peptides may be isolated therefrom by methods known in the art. The cells may also be administered to a subject, or alternatively, the cells may be modified directly in vivo.

The invention also relates to a combination comprising an antiviral peptide of the invention and a second agent selected from, for example, a nucleic acid, a peptide, a protein, a contrast agent, an antibody, a toxin, and a small molecule.

The invention also relates to recombinant libraries, e.g., phage libraries, comprising the peptides of the invention.

While the foregoing disclosure provides a general description of the subject matter included within the scope of the invention, including the methods of making and using the invention and the best mode thereof, the following examples are provided to further enable those skilled in the art to practice the invention and to provide a complete written description thereof. However, those skilled in the art will appreciate that the details of these examples are not to be construed as limitations of the present invention, the scope of which should be construed from the claims appended to this disclosure and their equivalents. Various other aspects and embodiments of the invention will be apparent to those skilled in the art in view of this disclosure. The details of these examples should not be considered as limiting.

As used herein, "and/or" should be considered a specific disclosure of each of the two specified features or components, with or without the other. For example, "a and/or B" will be considered a specific disclosure of each of (i) a, (ii) B, and (iii) a and B, as if they were individually listed herein.

Unless the context indicates otherwise, the description and definition of features set forth above is not limited to any particular aspect or embodiment of the invention, and applies equally to all aspects and embodiments described.

43页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:在糖尿病足的情况下增强组织氧合的医药制剂及其用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!