Alloferons - immunomodulatory peptides

Information

  • Patent Application
  • 20020151679
  • Publication Number
    20020151679
  • Date Filed
    December 27, 2000
    23 years ago
  • Date Published
    October 17, 2002
    21 years ago
Abstract
The invention belongs to the field of biologically active peptides specifically stimulating antiviral, antimicrobial and antitumor activity of the human and animal immune system.
Description


[0001] The present invention is concerned with immunomodulatory materials of natural origin. In particular, the present invention is concerned with peptides of invertebrate origin and pharmaceutical preparations comprising such peptides which are useful in the treatment of immune deficient conditions, infections and oncological diseases.


[0002] In the state of the art various pharmaceutical preparations of natural origin containing materials of animal, including insect, and plant tissues able to stimulate the immune system's efficacy are known.


[0003] A process for obtaining cellular protein having anti-HIV activity from CD4-positive T cells or myeloid cells is disclosed in U.S. Pat. No. 5,480,782.


[0004] A topic formulation comprising a Ginkgo biloba extract exhibiting antibacterial and antiviral properties is disclosed in DE 43 34 600 A1.


[0005] WO 96/04005 discloses a pharmaceutical composition for stimulation of the immune response of an organism comprising as the active ingredient major histocompatibility complex antigens extracted from animal tissues, serum or cells. The tissues, cells or sera are chosen from goat, veal or pig liver and bovine red blood cells.


[0006] A pharmaceutical composition containing an extract of the plant Nigella sativa is disclosed in U.S. Pat. No. 5,482,711 for treating cancer, preventing the side effects of anticancer chemotherapy, and for increasing the immune functions in humans.


[0007] WO 81/03124 discloses a polypeptide fraction isolated from the mussel Mytilus edulis and used as antibiotic composition effective against various viruses, bacteria and protozoa.


[0008] Antibacterial peptides from honey bees and a process for their isolation, production and applications have been disclosed in EP 0 299 828 A1.


[0009] Antibacterial peptides isolated from the Coleopteran insects, Tenebdo molitor and Leptinotarsa decemlineata are disclosed in WO 90/14098.


[0010] Antibacterial protein isolated from the Lepidopteran insect, Hyalophora gloveri is disclosed in EP 0 856 519 A2.


[0011] Antimicrobial peptides structurally similar with arginine-containing fragments of lentivirus transmembrane proteins are disclosed in U.S. Pat. No. 5,714,577.


[0012] Antiviral and antimicrobial peptides isolated from porcine leucocytes are disclosed in U.S. Pat. No. 5,804,558.


[0013] Immunomodulatory peptides specifically binding major histocompatibility complex class II antigens and decreasing in that way a possibility of autoimmune disease are disclosed in U.S. Pat. No. 5,827,516.


[0014] EP 0 320 528 A1 discloses the use of hemocyanins and arylphorins isolated from various molluscs and arthropods including the insect Calliphora erythrocephala as stimulants the production of specific antibodies and the antitumor activity of antibody-dependent T-lymphocytes.


[0015] The preparations mentioned above and analogous natural pharmaceutical preparations enhance the recent arsenal of medicines suitable for treatment of immune deficient conditions, infections and oncological diseases. However, the pharmaceuticals which are available up to now do not cover existing demands in immunomodulatory medicines.


[0016] Therefore, it is an object of the present invention to provide a pharmaceutical preparation having immunomodulatory activity and in particular being useful for the treatment of immune deficient conditions, infections and oncological diseases.


[0017] It has now surprisingly been found that specific peptides exhibit the desired immunomodulatory activity.


[0018] Thus, the present invention relates to a peptide consisting of up to 30 amino acid residues and having the following general structural formula (1):


X1-His-Gly-X2-His-Gly-Val-X3  (1)


[0019] wherein


[0020] X1 is absent or represents at least one amino acid residue,


[0021] X2 is a peptide bound or represents at least one amino acid residue, and


[0022] X3 is absent or represents at least one amino acid residue, or a pharmaceutically acceptable salt or ether thereof, the peptide exhibiting immunomodulatory activity.


[0023] The present invention provides a new class of immunomodulatory peptides, designated “alloferons” herein, representative members of which were isolated from the blood of bacteria challenged larvae of an insect, blow fly Calliphora vicina R.-D. (Diptera, Calliphoridae).


[0024] The alloferons of the invention have been found to stimulate cytotoxic anticancer activity of animal (mouse) and human natural killer cells. Experimental data on the alloferons' immunomodulatory activity show that they are able to stimulate the cytotoxic anticancer activity of human and mouse lymphocytes at extreme low concentrations. The minimum effective concentration was determined to be about 0.0005 nanogram/ml. The optimum concentration was found to be 0.05-0.5 nanogram/ml. Assuming the important role of natural cytotoxicity as effector mechanism of innate immunity (Trinchieri G., Advances in Immunology, 1989, vol. 47, 187-375; Brittenden J., Heys S. D., Ross J. and Eremin O., 1996, vol. 77, 1126-1243), alloferons may be useful as antiviral, antimicrobial and anticancer medicines of immunomodulatory mode of action.


[0025] Moreover, with regard to the stimulation of the anticancer activity of the cytotoxic lymphocytes, alloferons were found to induce intensive and prolonged interferon synthesis in experimental animals. Interferons are a group of key antiviral (alpha- and beta- interferons) and immunomodulatory (gamma-interferon) cytokins produced in the organism in response to viral infection and some other external stimuli. Elevation of interferons concentration in the blood helps to cure or mitigate a broad range of viral, oncological and autoimmune disorders. Injections of recombinant or natural interferons are successfully used in the immunotherapy of hepatitis C (Bekkering et al., J. Hepathology, 1998, 28, 6, p. 960-964), herpes (Cardamakis et al., Gynecol. Obstet. Invest., 1998, 46, 1, p.54-57), multiple myeloma (Zee et al., J. Clin. Oncol., 1998, 16, 8, p. 2834-2839), Hodgkin's disease (Aviles et al., Leuk. Lymphoma, 1998, 30,5-6, p. 651-656), myeloid leukemia (Gilbert H. S., Cancer, 1998, 83,6,p.1205-13), multiple sclerosis (Durelli et al., Mult. Scler, 1995,1, suppl.1, p. 32-37), atopic dermatitis (Schneider et al., Ann. Allergy Asthma Immunol., 1998, 80, 3, p. 263-268), fungal infections (Kullberg, Eur. J. Clin. Microbiol. Infect. Dis., 1997, 16, p. 51-55) etc. Moreover exogenic interferons, inducers of endogenic interferon synthesis such as bropirimine, a phenylpyrimidinone analog, might be used to achieve similar therapeutic results (Akaza et al., Eur. Urol., 1998, 34, p. 107-110).


[0026] Experimental data show that alloferons effectively induce interferon synthesis and stimulate some immunological reactions (natural killers activity) in a manner similar to interferons. Therefore alloferons are believed to have similar therapeutic use compared to interferons and interferon inducers including but not limited to treatment of interferon-sensitive viral and cancer diseases. Direct confirmation of this hypothesis is obtained in experiments with virus infected mice. It is shown that alloferon administration significantly increase the survival rate in mice intrapulmonary infected with a lethal dose of human influenza virus A and B.


[0027] The chemical structure of alloferons has no similarity with interferons, other known cytokines and interferon inducers as well as any other materials of medical importance. The chemical structure of alloferons and the mode of biological activity are also quite different of those of arylphorin isolated from Calliphora and demonstrating immunologic and antitumor activity as it is disclosed in EP 0 320 528 A1. Alloferons preferably have a molecular mass close to 1200 Da and belong to the unique peptide family which has not been described so far. Calliphora arylphorin has a molecular mass of about 500 000 Da (Naumann U. and Scheller K. Biochem. Biophys. Res. Communications, 1991, 177, p. 963-971) and is proposed to be used as an adjuvant in the course of specific vaccination and specific stimulation of antibody-dependent T-lymphocytes antitumor activity as it is disclosed in EP 0 320 528 A1. No data concerning a possible effect of arylphorin on the natural killer cell activity and interferon synthesis are available up to now.


[0028] Alloferons are linear peptides having a unique amino acid sequence represented by the general formula as follows:


X1-His-Gly-X2-His-Gly-Val-X3


[0029] where:


[0030] X1 is absent or represents at least one amino acid residue


[0031] X2 is a peptide bond or represents at least one amino acid residue, and


[0032] X3 is absent or represents at least one amino acid residue.


[0033] The alloferons of the present invention have up to 30, preferably up to 20 and most preferable 5-13 amino acid residues.


[0034] Examples of alloferons of the present invention are summarized in Table 1.
1TABLE 1Amino acid sequences of alloferons, the homologous to alloferon fragment ofinfluenza B virus precursor protein and general formula (1).PositionPeptide12345678910111213141516Alloferon 1HisGlyValSerGlyHisGlyGlnHisGlyValHisGlyAlloferon 2GlyValSerGlyHisGlyGlnHisGlyValHisGlyAlloferon 3ValSerGlyHisGlyGlnHisGlyValHisAlloferon 4SerGlyHisGlyGlnHisGlyValAlloferon 5ProSerLeuThrGlyHisGlyPheHisGlyValTyrAspAlloferon 6PheIleValSerAlaHisGlyAspHisGlyValAlloferon 7ThrHisGlyGlnHisGlyValAlloferon 8HisGlyHisGlyValHisGlyAlloferon 9LeuAlaSerLeuHisGlyGlnHisGlyValAlloferon 10CysValValThrGlyHisGlySerHisGlyValPheValAlloferon 11IleSerGlyHisGlyGlnHisGlyValProAlloferon 12CysGlyHisGlyAsnHisGlyValHisAlloferon 13IleValAlaArgIleHisGlyGlnAsnHisGlyValAlloferon 14HisGlySerAspGlyHisGlyValGlnHisGlyAlloferon 15PheGlyHisGlyHisGlyValAlloferon 16HisGlyAsnHisGlyValLeuAlaAlloferon 17HisGlyAspSerGlyHisGlyGlnHisGlyValAspAlloferon 18HisGlyHisGlyValProLeuAlloferon 19SerGlyHisGlyAlaValHisGlyValMetAlloferon 20TyrAlaMetSerGlyHisGlyHisGlyValPheIleInfluenza vi-HisGlyTyrThrSerHisGlyAlaHisGlyValrus B precur-sor (positions377-387)generalX1HisGlyX2HisGlyValX3formula (1)


[0035] Alloferons 1 and 2 are natural peptides isolated from the blood of bacteria challenged larvae of an insect, Calliphora vicina in the course of purposeful screening of cytokine-like materials able to stimulate cytotoxic activity of mammalian natural killer cells. Alloferons 3 and 4 are truncated forms of alloferon 1, which were chemically synthesized in order to determine possible biologically active modifications of the natural prototype molecule.


[0036] Comparative study of the effect of alloferon 1-4 on the cytotoxic activity of lymphocytes demonstrated that all of them are bioactive molecules. See Example 5. This makes possible to distinguish conservative (functionally important) and variable parts of the alloferon structure. Alloferons 5-20 are examples represented to show preferred modifications of variable fragments of the basic structure of alloferon.


[0037] A data base search did not reveal peptides of natural origin or bioactive synthetic peptides having close similarity to the alloferon structure. Therefore alloferons are believed to belong to a new family of bioactive peptides. Nevertheless, alloferons have, to certain extent, structural analogy with fragments of some functionally important proteins. For example, alloferon 1 has 63% identity with fragment 377-387 of the influenza virus B hemagglutinin precursor. Hemagglutinin is known to be a key membranotropic protein of the virus envelope responsible for the integration with the cell membrane of the host.


[0038] Alloferon 1 was used as a prototype molecule in the course of the development of the invention. Alloferon 1 is a linear peptide having a molecular mass of 1265 Da consisting of 13 amino acids. See Table 1. A comparison with alloferons 2-4 allows to determine functionally important elements of the structure of alloferon, which are necessary for its efficacy as a stimulant of NK cell's cytotoxicity and, other activities, and to predict possible structural modifications, which do not change the biological activity of the peptide.


[0039] Comparison of alloferon 1 with the structure of alloferons 2-4 shows that the presence of the fragment Ser-Gly-His-Gly-Gln-His-Gly-Val is sufficient to conserve the biological activity as since all the peptides exhibit similar activities in NK cell cytotoxicity test. Therefore, this fragment or a part of the fragment represents the core conservative structure in alloferon sequences. Positions 1-3 in the alloferon 1 molecule can be missing or can be replaced by one or more amino acids.


[0040] Furthermore, a comparison with the homologous fragment of the influenza virus hemagglutinin reveals that positions 4 and 5, represented in the alloferon 1 sequence by amino acids serin (Ser) and glycin (Gly), can be replaced by some other amino acid preferably chosen from the group of aliphatic, aromatic or heterocyclic amino acids. For instance, serin can be replaced by threonin (Thr) and glycin by serin.


[0041] Thus, the available data reveals that the first five amino acids in the alloferon 1 sequence are a variable fragment which can be absent or contain at least one amino acid. Therefore, this fragment is marked in the alloferon structural formula (1) as X1. Advantageously, X1 is selected from the group consisting of nothing, His-Gly-Val-Ser-Gly-, Gly-Val-Ser-Gly-, Val-Ser-Gly-, Ser-Gly-, Pro-Ser-Leu-Thr-Gly-, Phe-Ile-Val-Ser-Ala-, Thr-, Leu-Ala-Ser-Leu-, Cys-Val-Val-Thr-Gly-, Ile-Ser-Gly-, Cys-Gly-, Ile-Val-Ala-Arg-Ile-, Phe-Gly-, His-Gly-Asp-Ser-Gly-, Ser-Gly- and Tyr-Ala-Met-Ser-Gly-.


[0042] Similarly, positions 14-15 in the alloferon 1 molecule can be missing or can be replaced by a sequence of one or more amino acids. Therefore, this fragment is marked in the alloferon structural formula (1) as X3. Advantageously, X3 is selected from the group consisting of nothing , -His-Gly, -His, -Tyr-Asp, -Phe-Val, -Pro, -Gln-His-Gly, -Leu-Ala, -Asp, -Pro-Leu, -Met and -Phe-Ile.


[0043] Moreover, comparison of alloferon and the corresponding hemagglutinin fragment reveals that position 9, occupied in the alloferon molecule by glutamin, is also variable and glutamin can be replaced by some other amino acid, for example, by alanin. Consequently, position 9 of the alloferon structural formula (1) is marked as X2,which can be a peptide bond linking Gly and His or contain not less then 1 amino acid, preferably 0-3 amino acids, more preferably 0-2 amino acids and most preferable 1 amino acid, in particular -Gin-.


[0044] Advantageously, X2 is selected from the group consisting of a peptide bond, -GIn-, -Phe-, -Asp-, -Ser-, -Asn-, -Ala-, -Gln-Asn-, -Ala-Val- and -Ser-Asp-Gly-.


[0045] Incorporation of the alloferon sequence into a larger molecule such as a carrier protein without significant alteration of the biological activity of the alloferon is also possible. Thus, the present invention also relates to chemical compounds such as peptides or proteins comprising an amino acid sequence having the above general formula (1), provided that the peptide or protein is not naturally occurring, and in particular not the influenza virus B precursor.


[0046] Complex immunological, pharmacological and toxicological studies summarized in the examples below demonstrate a range of useful properties of alloferons. The obtained data show that alloferon is a new cytokine-like peptide. The mode of action of alloferon comprises stimulation of nonself or aberrant self recognition of cells and lysis by the cytotoxic lymphocytes as well as induction of interferon synthesis. Therefore, alloferon is useful as immunomodulatory medicine to correct a deficiency in the production of interferons and activity of natural killer cells, treatment of viral, oncological and other diseases dependent on the said deficiency. Alloferon is practically nontoxic, has no teratogenic, embryotoxic or mutagenic properties as it is shown in advanced preclinical studies.


[0047] The experimentally established properties of alloferon 1 are summarized in Table 2.
2TABLE 2Experimentally proved pharmacological activities of alloferon 1.Effective in vivo dose(mg/kg body mass) orin vitro concentrationActivity(ng/ml)Medical use1. Stimulation of mouse0.05-50ng/mlTherapy of infectiousspleen lymphocytes'and oncologicalcytotoxic activitydiseases2. Stimulation of mice'1.25mg/kgInfluenza therapyresistance to theinfluenza virus Ainfection3. Stimulation of mice'1.25mg/kgInfluenza therapyresistance to theinfluenza virus Binfection4. Interferon synthesis0.125-1.25mg/kgTherapy andinduction in miceprophylaxis ofviral and oncologicaldiseases5. Stimulation of human0.0005-500ng/mlTherapy andperipheral bloodprophylaxis oflymphocytes' cytotoxicviral and oncologicalactivitydiseases6. Stimulation of5ng/mlAdjuvant therapyperipheral bloodof cancerlymphocytes' cytotoxicactivity in cancer patients


[0048] The pharmacological activity spectrum, in general, corresponds to known properties of interferon-alpha concerning the influence on the cytotoxic activity of natural killer cells and antiviral resistance. In that aspect, alloferon can be characterized as interferon-alpha functional analog. The mode of action of alloferon, regarding in vitro stimulation of NK cells cytotoxic activity, is observed at very low concentrations—about 1 picogram/mI (10−9 g/ml). In that aspect alloferon is as active or more active as endogenous cytokines, interferons and interleukins.


[0049] Moreover, alloferon is able to induce, alone or in cooperation with interleukin 12, the synthesis of endogenic interferons, including interferon-gamma. Therefore, alloferon can be attributed to the group of interferon inducers.


[0050] Preclinical studies of the in vivo activity of alloferon show that it has potent antiviral activity when tested using as a model mice infected by human influenza virus. In this model wild type males were infected intranasally by a suspension of the human influenza virus and alloferon 1 was injected intraperitoneally one day before infection and then 1, 2, 4, 6 and 8 days after. Alloferon effectively protected mice from pulmonary lesions and death. Thus, alloferons are useful in the preparation of a pharmaceutical preparation for the treatment or prevention of viral infections.


[0051] Neither an acute nor a chronic toxicity of alloferon 1 was found in the course of in vivo and in vitro studies.


[0052] It is understood that the pharmaceutical preparations of the present invention may also comprise conventional additives like excipients or carriers. The preparations may be administered to the patient by intranasal, enteral, such as oral or rectal, and parenteral, such as intraperitoneal, intramuscular, intravenous or subcutaneous route. The preparations may be administered in dosage forms such as intranasal dropping solutions, sprays, liposomes, capsules, tablets and suppositories. For parenteral use the pharmaceutically active components are preferably in the form of an injectable solution.


[0053] Thus, alloferons are useful in the treatment or prophylaxis of various infectious or oncological diseases where improvement of innate immunity, including interferon system and natural cell mediated cytotoxicity, can have therapeutic significance. The examples of conditions under which alloferons application is prospective comprise influenza virus and other respiratory viral infections, viral hepatitis, AIDS and AIDS relevant secondary infections and oncological conditions, acute and chronic leukemia and other cancers where interferon treatment efficacy is proved, fungal systemic infections sensitive to the interferon treatment etc.


[0054] In spite of a certain similarity in biological activity (NK cell cytotoxic activity stimulation, indirect antiviral activity), alloferons differ very much of interferons in terms of structure and mode of action. Thus, interferons are glycoproteins with molecular mass ranging from 17 000 to 80 000 daltons. Glycosilation of amino acid chain is a necessary condition of interferon functional activity as well as their tissue and species specificity. Alloferon is a preferably nonglycosilated oligopeptide having molecular mass of preferably about 1265 Da, 13-60 times less then interferons molecular masses. The amino acid sequence of alloferon has no similarity with any fragment of interferon sequences. There are essential differences between alloferons and interferons in functional aspect as well. Thus, alloferon induces the production of endogenic interferons and promotes in this way a cascade of defense responses mediated by interferons. Exogenic interferon application may rather suppress endogenic interferon synthesis by means of negative feedback mechanism.


[0055] The peptides of the present invention can be isolated from natural sources or synthesized by known methods. The peptides of this invention can also be produced by recombinant DNA techniques. Thus, the invention comprises cultivating a cell host previously transformed with a suitable vector containing a DNA sequence, e.g. a cDNA encoding a peptide sequence including any of the peptides of this invention, said DNA sequence being placed under the control of a promoter and followed by termination signals recognized by the cell host machinery such as to authorize the expression said DNA sequence of said peptide sequence, and recovering the peptide sought from the expression of products of the cell culture. Advantageous host cells belong to Lactobacillus strains, E. coli, Agrobacterium or Bacillus strains. Alternatively the peptides can be easily produced by well known chemical synthesis.






[0056] The invention is further illustrated by the following examples:


Example 1

[0057] Isolation of alloferons from insect blood, structural characterization and chemical synthesis


[0058] Alloferons were initially discovered in the blood of the immunized (bacteria challenged) insect, blowfly Calliphora vicina. Postfeeding C. vicina larvae maintained in the laboratory conditions as described (Chernysh S.l., Simonenko N. P., Numata H. Appl. Entomol. Zool., 1995, Vol. 30, No. 3, p. 498-499) were bacteria challenged by the pricking off cuticle with a needle soaked in a suspension of heat-killed Escherichia coli and Micrococcus luteus cells. The hemolymph of septically injured larvae was collected, centrifuged and applied onto a Sep-Pak C18 chromatographic column (Waters Co). The column was washed with 0.05% trifluoroacetic acid. Then the target materials were eluted with 50% acetonitril acidified with 0.05% trifluoroacetic acid. The eluted composition was lyophilized and used to stepwise chromatographic purification of the active principle. The biological activity of the fractions was monitored during purification steps using mouse spleen lymphocytes as cytotoxic cells and H3-uridine labeled K562 cancer cells as a target.


[0059] As a result of the purification steps, two close oligopeptides demonstrated potent immunomodulatory activity and referred to as alloferons 1 and 2 were isolated from the primary composition and chemically characterized. The amino acid sequence of the peptides was determined using an automated Edman degradation method on a model 473A sequenator (Applied Biosystems). The structure of alloferon 1 and 2 was determined as follows:


His -Gly-Val-Ser-Gly-His -Gly-GIn-His -Gly-Val-His -Gly (alloferon1)


Gly-Val-Ser-Gly-His -Gly-GIn-His -Gly-Val-His -Gly (alloferon 2)


[0060] The peptides were further analyzed by MALDI-TOF ionozation mass spectrometry on a Bruker (Bremen) BIFLEX matrix-assisted laser desorption time-of-flight mass spectrometer and their molecular masses were experimentally determined as 1265 Da (alloferon 1) and 1126 Da (alloferon 2). The masses of alloferon 1 and 2 deduced from the amino acid sequencing data and the masses determined by mass spectrometry are in good agreement confirming that alloferon 1 and 2 are linear peptides having no posttranslational modifications. Alloferon 1 was selected as a prototype molecule for biological and preclinical studies. In order to get sufficient amount of alloferon 1 for the further experimentation, it was chemically synthesized by means of solid phase peptide synthesis technology (Neimark J. and J. P. Brian, Peptide Research, 1993, vol.6, p. 219) The peptide purification protocol included two major steps. First step was performed on the Sep-Pak Vac columns with C18 sorbent (Waters) by means of the column elution by 40% acetonitril acidified by 0.05% trifluoroacetic acid. Finally the peptide was purified to homogeneity using a Beckman Gold System chromatograph equipped with an Aquapore ODS Prep 10 C18 (100×10 mm, Brownlee) column in the linear gradient of 0.05% trifluoroacetic acid and acidified acetonitril (0-20% acetonitril during 40 min under flow rate 2.5 ml/min and detector wave length 225 nm). The peptide purity was confirmed by MALDI-TOF mass spectrometry. The amino acid sequence's accuracy was confirmed by microsequencing.


[0061] Truncated forms of alloferon 1, alloferons 3 and 4, were synthesized, purified and controlled in the same way as described for alloferon 1.



Example 2

[0062] Effect of alloferon 1 on the cytotoxic activity of mouse spleen lymphocytes


[0063] To analyze the effect of alloferon on the mouse spleen lymphocytes' cytotoxic activity, the standard cytotoxicity assay was used (Hashimoto J. and Sudo E., Gann, 1971, vol. 62, 139-145; Filatova N. A., Malygin A. M., Goryunova L. B., Fel V. Ya. and Khavinson V. K., Tsitologia,m 1990, Vol. 32, No. 6,652-658). H3-uridine labeled K562 human leukemia cells were used as targets for a cytotoxic lymphocytes' attack. Fresh spleen lymphocytes and target cells were co-incubated during 18 hours in the presence or absence of the preparation. Then the proportion of killed and normal target cells and the corresponding cytotoxicity indices were determined in control and experimental groups.


[0064] A typical result of the synthetic alloferon 1 stimulatory activity is shown in Table 3.


[0065] Alloferon administration to the incubation medium in a broad concentrations range (0.05-50 ng/ml) induced statistically significant amplification of the cytotoxic activity of natural killer cells against target tumor cells. A concentration of 500 ng/ml was not stimulatory. However, in that case the cytotoxic activity was not suppressed below the control level as well.


[0066] Therefore, even 10000-times excess of the minimum effective concentration was not harmful to the cytotoxic activity of NK cells.


[0067] Thus, alloferon efficiently stimulates the cytotoxic activity of mouse spleen lymphocytes at very low concentrations characteristic to specific cytokines such as interferon-alpha or interleukin 2 responsible for NK cells activation. At the same time, alloferon did not demonstrate immunosuppressive properties even under 10000-fold excess of the effective concentration.
3TABLE 3in vitro effect of alloferon 1 on the mouse spleenlymphocytes' cytotoxicity to K562 human leukemia cellsConcentrationCytotoxicity indexTreatmentng/mlAverage, % (n = 18)% to controlControl021.3 ± 3.0 100Alloferon0.05 35.2 ± 4.0**1650.5 39.3 ± 3.9***1855 34.3 ± 4.5**16150 37.2 ± 4.5**17550020.3 ± 3.6  95**P < 0.01; ***P < 0.001



Example 3

[0068] Effect of alloferon 1 on the cytotoxic activity of human peripheral blood lymphocytes


[0069] The determination of the cytotoxicity index has been performed as described in Example 2. Human peripheral blood lymphocytes were released from fresh donor blood and purified of erythrocytes by centrifugation using the histopak 1077 solution (Sigma). After centrifugation the lymphocytes were resuspended in phosphate buffer, centrifuged and resuspended again in RPMI 1640 medium supplemented with RNAase. The lymphocytes were diluted up to 2×106 cells/ml and immediately used for the cytotoxicity analysis. Interferon-alpha 2b (Intron, Shering-Plough), a natural stimulant of NK cells' cytotoxic activity was used as a positive control.


[0070] The PBLs' cytotoxicity against K562 cancer cells was significantly increased when the preparation was added to the incubation medium in a concentration starting from 0.0005 nanogram/ml, however, the stimulatory activity reached a plato at a concentration of about 0.05 nanogram/ml (Table 4). Interferon-alpha 2b administered in a concentration of 5 ng/ml was less effective as compared to the alloferon administered in the optimal concentration rang of 0.05-0.5 ng/ml.


[0071] These experimental data demonstrate a strong stimulatory effect of alloferon on the in vitro cytotoxic activity of human peripheral blood lymphocytes directed to the lysis of tumor cells.
4TABLE 4Effect of alloferon and inteferon-alpha 2b on thecytotoxicity of human periferal blood lymphocytes.ConcentrationCytotoxicity indexTreatmentng/ml%% to controlControl027.3 ± 7.3100  Inteferon-alpha 2b564.3 ± 3.8236***Alloferon0.000562.0 ± 6.4227***0.00573.8 ± 1.7270***0.0579.8 ± 5.0292***0.579.8 ± 2.8292***566.8 ± 7.2245***5068.0 ± 5.3249***50068.8 ± 4.4252******P < 0.001



Example 4

[0072] Comparative study of alloferon I and interferon-alpha 2b efficacy variation regarding stimulation of the cytotoxic activity of lymphocytes in the population of healthy donors


[0073] PBLs' cytotoxic activity was monitored in the random sampling of 17 healthy donors in order to evaluate diversity and correlation of the lymphocytes' responses to alloferon and interferon-alpha 2b stimulation. The cytotoxic activity was evaluated as described in Examples 2 and 3. Two kinds of target cells were used in the study simultaneously: the K562 cell line originated from erythromyeloid leukemia cells and the A431 cell line originated from the solid colorectal tumor. The proportion of lymphocytes and target cells (E/T ratio) in the trial was 20:1.


[0074] The data obtained are summarized in Table 5. Each figure in the Table comprises results of 6 cytotoxicity determinations. The data show significant variation of the cytotoxic activity of the lymphocytes in the control groups, particularly a different capacity to recognize and eliminate the target cells of different origin. Responses to the preparation's administration are also different. Nevertheless a clear correlation of the responses to the alloferon and interferon administration was found in most donors: donors positively responding to interferon in most cases also positively respond to alloferon and vice versa.


[0075] Generalized figures of the efficacy of two preparations calculated on the basis of the data in Table 5 are shown in Table 6. Most donors were responding to interferon (10 out of 17 donors) and alloferon (also 10 donors) by statistically approved increase of the cytotoxic activity against K562 or A431 targets. Responsivenesses to alloferon and interferon administration were in good correlation: 9 donors were equally responsive to both preparations and each one donor was selectively responsive to interferon or alloferon, respectively.


[0076] Thus, an analysis of the healthy donors' responsiveness shows that alloferon and interferon-alpha 2b possess similar efficacy in this model. Moreover, two preparations seem interchangeable as stimulants of the activity of cytotoxic lymphocytes in most although not all individuals.
5TABLE 5Effect of alloferon and interferon-alpha 2b (Kfin = 5 ng/ml) on thecytotoxicity of peripheral blood lymphocytes (PBL) in healthydonors (lymphocyte : target ratio = 20:1).Cytotoxicity index,Statistically significantPrepara-M ± m, %stimulation (P ≦ 0.05)tionK562A431K562A4311Control5.3 ± 4.453.8 ± 6.5Interferon50.2 ± 1.6***55.7 ± 8.6+Alloferon30.2 ± 5.5**83.7 ± 2.9**++2Control14.2 ± 3.3−64.5 ± 40.6Interferon50.2 ± 2.5***−88.3 ± 40.4+Alloferon39.8 ± 1.5***−86.7 ± 22.8+3Control29.8 ± 7.4−53.6 ± 19.9Interferon21.8 ± 7.527.0 ± 6.5**+Alloferon30.8 ± 6.839.7 ± 3.3***+4Control30.0 ± 2.983.8 ± 3.6Interferon42.8 ± 2.4**89.5 ± 2.3+Alloferon45.4 ± 2.3***91.2 ± 0.9+5Control43.3 ± 3.5−14.8 ± 8.3Interferon64.2 ± 57***35.4 ± 5.2***++Alloferon51.0 ± 1.724.4 ± 4.0**+6Control41.7 ± 10.550.0 ± 5.7Interferon30.5 ± 8.643.7 ± 5.0Alloferon37.6 ± 10.253.7 ± 4.47Control40.3 ± 3.217.3 ± 5.5Interferon58.2 ± 2.5**11.8 ± 8.3+Alloferon59.2 ± 5.2**−10.2 ± 8.0+8Control23.0 ± 3.5−20.0 ± 6.3Interferon19.2 ± 2.374.0 ± 2.7***+Alloferon29.2 ± 6.380.7 ± 1.8***+9Control6.2 ± 11.37.8 ± 15.6Interferon25.0 ± 4.638.5 ± 3.4Alloferon25.0 ± 12.324.5 ± 15.110Control34.3 ± 1.374.0 ± 4.5Interferon51.8 ± 2.9**82.7 ± 1.9+Alloferon44.8 ± 3.5*88.4 ± 1.2**++11Control57.0 ± 4.437.2 ± 6.7Interferon52.0 ± 2.250.8 ± 9.2Alloferon42.8 ± 5.946.5 ± 7.612Control61.8 ± 3.455.6 ± 4.9Interferon71.8 ± 2.9*51.4 ± 3.6+Alloferon47.8 ± 4.156.4 ± 3.113Control44.0 ± 13.152.8 ± 6.5Interferon62.7 ± 7.558.2 ± 6.1Alloferon49.7 ± 3.954.8 ± 4.014Control30.2 ± 6.1−6.0 ± 8.1Interferon14.7 ± 8.748.7 ± 4.5**+Alloferon2.0 ± 4.249.8 ± 12.9**+15Control16.8 ± 2.311.2 ± 6.8Interferon2.2 ± 3.825.0 ± 5.7Alloferon19.5 ± 6.827.7 ± 3.716Control−3.5 ± 7.461.4 ± 7.1Interferon5.0 ± 4.769.2 ± 2.1Alloferon18.5 ± 1.7*68.7 ± 2.0+17Control23.3 ± 5.657.8 ± 4.1Interferon29.0 ± 3.362.5 ± 1.7Alloferon24.7 ± 4.064.3 ± 4.1*P < 0.05; **P < 0.01; ***P < 0.001


[0077]

6





TABLE 6










Comparative characteristics of the efficacy of alloferon and


interferon-alpha 2b regarding PBLs' cytotoxic activity stimulation


in the random sampling of healthy donors (from the data of Table 5).









Indicatior
Interferon
Alloferon











Number of donors
17









Positive responses* proportion




(taking into account target specificity):


K562
 7/17 = 41%
 6/17 = 35%


A431
 4/17 = 24%
 6/17 = 35%


K562 or A531
10/17 = 59%
10/17 = 59%


Positive responses* coincidence:








Interferon and alloferon sensitive donors
9/17 = 53%


Donors selectively sensitive to interferon
1/17 = 6%


Donors selectively sensitive to alloferon
1/17 = 6%






*Positive response = statistically significant increase of lymphocyte cytotoxicity index









Example 4

[0078] Comparative study of the efficacy variation of alloferon 1 and interferon-alpha 2b regarding stimulation of the lymphocytes' cytotoxic activity in the random sampling of cancer patients.


[0079] Blood samples from 18 patients with different malignancies have been tested according to the same protocol as healthy donors of Example 3. Results are shown in Table 7. Positive responses to alloferon treatment were registered in various patient groups, particularly those suffered by chronic and acute leucosis (5 of 9 cases). Positive responses were also detected in patients suffered by non-Hodgkin lymphoma (2 of 6 cases) and one lung cancer patient.


[0080] The proportion of individuals positively responding to interferon or ailoferon was slightly decreased in cancer patients compared to healthy donors but also significant (56% and 50% of the total number, respectively). The correlation of responsiveness to interferon and alloferon was also weaker compared to healthy donors although evidential as well. According to the given test, alloferon seems to be an adequate replacement of injectable interferon in some part of cancer patients. However, assuming that alloferon not only mimics interferon activity but also stimulates the production of endogenic interferons, alloferon appears to be even more prospective as replacement of injectable interferon because of the expected double stimulation of the immune response via direct activation of natural cytotoxicity and induction of endogenic interferon synthesis.
7TABLE 7Effect of alloferon and interferon-alpha 2b on the cytotoxicity ofperipheral blood lymphocytes (PBL) in cancer patients(lymphocyte:target ratio = 20:1).StatisticallysignificantstimulationDiag-Cytotoxicity index, %(P ≦ 0.05)nosisTreatmentK562A431K562A4311ChronicControl11.2 ± 10.6−4.2 ± 8.7leucosisInterferon−5.5 ± 7.911.7 ± 12.5Alloferon−35.3 ± 5.4−9.0 ± 12.72ChronicControl4.8 ± 5.437.0 ± 5.6leucosisInterferon−10.7 ± 4.933.3 ± 9.0Alloferon1.3 ± 2.38.2 ± 8.33ChronicControl−8.3 ± 2.3−16.2 ± 10.6leucosisInterferon−3.5 ± 4.033.5 ± 7.4**+Alloferon−12.8 ± 3.726.2 ± 3.8**+4ChronicControl−11.0 ± 3.628.2 ± 5.9leucosisInterferon−9.2 ± 7.018.8 ± 4.3Alloferon−1.5 ± 5.119.2 ± 6.35ChronicControl−23.0 ± 5.315.2 ± 8.3leucosisInterferon−4.6 ± 9.646.0 ± 4.0**+Alloferon−8.0 ± 18.842.5 ± 3.9**+6AcuteControl42.3 ± 2.76.2 ± 16.5leucosisInterferon25.2 ± 4.0**43.5 ± 6.4*+Alloferon20.2 ± 6.8**48.5 ± 13.5*+7AcuteControl29.2 ± 5.6−18.7 ± 14.6leucosisInterferon50.2 ± 2.3**24.8 ± 10.1*++Alloferon28.2 ± 2.620.0 ± 6.9*+8AcuteControl15.2 ± 9.425.2 ± 6.5leucosisInterferon40.0 ± 3.4**17.8 ± 8.3+Alloferon23.5 ± 5.540.4 ± 6.29AcuteControl23.5 ± 8.2−4.8 ± 9.0leucosisInterferon25.1 ± 4.651.5 ± 6.5***+Alloferon13.8 ± 4.451.8 ± 4.8***+10LungControl11.5 ± 3.7−7.2 ± 10.2cancerInterferon27.2 ± 4.2**59.5 ± 3.9***++Alloferon−0.3 ± 6.155.5 ± 2.7***+11UterusControl30.5 ± 1.393.3 ± 1.0cancerInterferon50.8 ± 4.9***93.5 ± 0.9+Alloferon24.0 ± 4.494.3 ± 0.412Hodg-Control22.2 ± 8.790.3 ± 0.95kinInterferon39.3 ± 4.995.2 ± 0.79**+lym-Alloferon27.7 ± 4.890.2 ± 0.47phoma13Non-Control11.5 ± 6.482.8 ± 1.1Hodg-Interferon13.8 ± 3.080.2 ± 2.0kinAlloferon17.7 ± 2.891.5 ± 0.8***+lym-phoma14Non-Control16.3 ± 9.6−40.0 ± 20.5Hodg-Interferon31.2 ± 9.226.0 ± 8.9**+kinAlloferon36.7 ± 9.115.3 ± 4.8**+lym-phoma15Non-Control19.2 ± 12.157.7 ± 4.4Hodg-Interferon47.5 ± 6.6*62.2 ± 5.2+kinAlloferon37.3 ± 4.764.0 ± 5.0lym-phoma16Non-Control35.8 ± 8.347.2 ± 10.4Hodg-Interferon43.2 ± 3.543.5 ± 4.4kinAlloferon37.3 ± 7.056.3 ± 4.4lym-phoma17Non-Control49.4 ± 3.266.7 ± 6.2Hodg-Interferon48.8 ± 3.567.0 ± 3.3kinAlloferon68.3 ± 4.6**64.3 ± 5.1+lym-phoma18Non- Control6.3 ± 14.929.8 ± 4.6Hodg-Interferon−3.0 ± 8.733.2 ± 7.4kinAlloferon−10.7 ± 14.132.8 ± 9.2lym-phoma*P < 0.05; **P < 0.01; ***P < 0.001


[0081]

8





TABLE 7










Comparative characteristics of the efficacy of alloferon and


interferon-alpha 2b regarding PBLs' cytotoxic activity stimulation


in the random sampling of cancer patients (from the data of Table 7).









Indicatior
Interferon
Alloferon











Number of donors
18









Positive responses* proportion




(taking into account target specificity):


K562
 6/18 = 33%
1/18 = 6%


A431
 8/18 = 44%
8/18 = 44%


K562 or A531
10/18 = 56%
9/18 = 50%


Positive responses* coincidence:








Interferon and alloferon sensitive donors
7/18 = 38%


Donors selectively sensitive to interferon
4/18 = 22%


Donors selectively sensitive to alloferon
2/18 = 11%






*Positive response = statistically significant increase of lymphocyte cytotoxicity index









Example 5

[0082] Influence of alloferon I structural analogs on the cytotoxic activity of human peripheral blood lymphocytes


[0083] The activity of alloferon 1 analogs, alloferons 3 and 4, was investigated according to the protocols described in Examples 2 and 3. The mononuclear fraction isolated from the blood of healthy donors and target cell of the A431 cell line were co-incubated in the presence of one of the preparations: alloferon 1, alloferon 3, alloferon 4 or interferon-alpha 2b (positive control). The preparation's concentration in all cases was 5 ng/ml. Cytotoxicity index excess over the untreated control was used as the efficacy criterion.


[0084] The data of Table 9 demonstrate similar efficacy of alloferons 3 and 4 compared to alloferon 1 and interferon-alpha 2b.


[0085] Thus, the comparative analysis of the efficacy of alloferon 1 structural analogs, alloferons 3 and 4, shows that positions 1-4 and/or 14-15 in the alloferon 1 amino acid sequence (See Table 1) are unnecessary for it's specific pharmacological activity and, therefore, represent variable parts of the alloferon 1 structure which can be changed or replaced without activity loss.
9TABLE 9Effect of alloferons 1, 3 and 4 and interferon-alpha 2bon the cytotoxic activity of human peripheral bloodlymphocytes against A431 tumor cellsExperimentsAverage cytotoxicity index,PreparationnumberM ± m, %PControl6−6.0 ± 8.1Interferon648.7 ± 4.6<0.001Alloferon 14 49.8 ± 12.8<0.01Alloferon 3660.2 ± 2.7<0.001Alloferon 4560.8 ± 2.4<0.001



Example 6

[0086] in vivo antiviral activity of alloferon on mice infected by human influenza virus A


[0087] The antiviral activity of alloferon was investigated using the model of lethal infection of mice by the human influenza virus A. A suspension of the pathogenic to mice virus strain A/Aichi/2/68 (cerotype H3N2) was administered intranasally in a dose equal to 10 LD50 doses to wild type males with body mass ranging from 20 to 22 g. Alloferon 1 dissolved in 0.5 ml of 0.9% NaCl was injected intraperitoneally one day before virus inoculation, then 1, 2, 4, 6 and 8 days after inoculation. The preparation was tested in two doses: 25 and 2.5 microgram per mouse (0.5 and 0.05 microgram/kg). Control mice were injected with an equal volume of the solvent. The mortality of the mice was monitored during 10 days after infection.


[0088] The data of Table 10 show significant decrease of post infection mortality in the group treated with 25 micrograms of alloferon. The 2.5 microgram dose was not effective.
10TABLE 10Alloferon antiviral activity in mice inoculated by human influenza virus ADosage,Mortality 10 days aftermicrogramvirus inoculationTreatmentper mouseNN%Control201470Alloferon2,5201365Alloferon2520 5 25**P < 0.05


[0089] Similar results were obtained in the experiment where the antiviral activity of alloferon was compared with that of remantadin. See Table 11. Remantadin (amantadine derivative) is one of the most powerful antiviral agents specifically effective against influenza virus A (Ershov F. I. Antiviral preparations, Medicina, Moscow, 1998, 187 pp). Alloferon was injected subcutaneously in a dose of 25 microgram one day before virus inoculation, then one hour before inoculation, then 1 and 2 days post infection. Remantadin in the dose 1000 microgram was given per os one day before virus inoculation, then one hour before inoculation, then 1, 2 and 3 days post infection.


[0090] Both alloferon and remantadin effectively protected most of the infected animals from lethal pulmonary lesions caused by the influenza virus. Remantadin appears to be slightly more effective in the case of influenza virus A infection, however it must be used in a significantly larger dosage (about 40 times higher) compared to the alloferon. Moreover, remantadin is known to be ineffective in the case of influenza virus B and other viral infections contrary to alloferon which is equally effective to A and B strains of the influenza virus.
11TABLE 11Antiviral activity of alloferon and remantadin in miceinfected with human influenza virus ADosage,Mortality 10 days aftermicrogramvirus inoculationTreatmentper mouseNN%Control201365Alloferon2518 4 22*Remantadin100019 1  5****P < 0.05



Example 7

[0091] in vivo antiviral activity of alloferon on mice infected by human influenza virus B


[0092] The anti-virus B efficacy of alloferon was tested according to the protocol described in Example 6. Animals were intranasally inoculated with pathogenic to mouse Lee {fraction (1/40)} human influenza virus B strain in doses equal to 3 and 30 LD50. Ribavirin, 1-beta-D-ribofuranosyl- 1,2,4,-triazole-3-carboxamide, an antiviral agent effective against various influenza virus strains (Liao H. J. and Stollar V. Antiviral Res., 1993, 22, 285; Ershov F. I. Antiviral preparations, Medicina, Moscow, 1998, 187 pp) was used as a positive control.


[0093] The results are shown in Table 12. The infection caused severe pneumonia with high mortality rate in both control groups, independent of the virus dosage. Ribavirin effectively protected mice inoculated with a lower virus dose (3 LD50), however it was not effective against a higher virus dose (30 LD50). At the same time, alloferon was equally effective in both cases. Therefore alloferon demonstrated better antiviral efficacy compared to the known antiviral agent, ribavirin. Moreover, alloferon is effective at a dose approximately 10 times less of ribavirin's therapeutic dose.
12TABLE 12Antiviral activity of alloferon and ribovirin in miceinfected with human influenza virus BVirus doseMortality 10 days after(LD50Animalsvirus inoculationTreatmentequivalents)numberNumber%Control301310 77 310880Ribavirin,3010660250 microgram 3100  0***Alloferon,30102 20**25 microgram 3100  0*****P < 0.01 ***P < 0.001



Example 8

[0094] in vivo effects of alloferon on the interferon synthesis in mice


[0095] In order to investigate the possible mode of alloferon's antiviral activity, the in vivo effect of the preparation on the interferon synthesis in mice was studied. The interferon concentration in the blood serum of alloferon treated and control (untreated) animals was determined using as a model a monolayer of L-929 cells. Vesicular stomatitis virus (Indiana strain) has been used as a test-virus in a dose 100 times exceeding the 50% cytopathogenic doses. One unit of interferon activity is expressed as a value reciprocal to the mouse serum dilution protecting 50% L-929 cells against the cytotoxicity of the test- virus. Cycloferon was used as a positive control. Cycloferon is an interferon inducer which belongs to the chemical group of acridanons (Ershov F. I. Antiviral preparations, Medicina, Moscow, 1998, 187 pp). Alloferon or cycloferon were injected intraperitoneally in a dose of 25 microgram and 500 microgram, respectively.


[0096] The results of two series of the experiment are summarized in Table 13. In each series the experimental groups comprised 4 animals. Blood samples were collected from each animal, then individual serum aliquots were combined and used for the interferon determination. Alloferon stimulated statistically significant the growth of the interferon concentration with a maximum efficacy reached 24 h post treatment.
13TABLE 13Effect of alloferon on the interferon synthesis in mice.AnimalsExperimentsInterferon titre,TreatmentHoursnumbernumberArbitrary unitsControl815 ± 5 Cycloferon, 48295 ± 45500 microgram2482  49 ± 3.8*Alloferon, 282  31 ± 13.825 microgram 482 21 ± 3.72482  71 ± 16.2**P < 0.05


[0097] Another example of the in vivo effect of alloferon on the interferon synthesis in mice is shown in the Table 14. The method was the same as in the previous experiment except that alloferon was administered subcutaneously and individual blood samples were analyzed separately. Both alloferon and cycloferon stimulated the interferon production in short-term prospect, 6 hours after treatment. During 24 hours the titre of interferon returned to the control level both in alloferon and cycloferon treated animals.


[0098] Thus, the data above confirm that alloferon injection has an in vivo interferon inducing activity on the level similar to those of cycloferon, a known interferon inducer. The length of the stimulatory effect can range from 6 to 24 hours or more, depending, probably, on the physiological state of the animal and way of administration. Compared to traditional chemical interferon inducers such as cycloferon alloferon has the advantage to be effective at a much lower dosage (about 20 times less).
14TABLE 14Short-term effect of alloferon on the interferon synthesis in mice.Time,AnimalsInterferon titer,TreatmenthnumberArbitrary unitsControl77.1 ± 1.8 Cycloferon, 6732.9 ± 10.7*500 microgram24711.4 ± 2.8 Alloferon, 6840.6 ± 11.4*25 microgram24814.4 ± 4.3 *P < 0.05



Example 9

[0099] Alloferon toxicity evaluation


[0100] The toxicity of alloferon was tested using a panel of in vitro and in vivo models. See Table 15. No signs of acute or chronic toxicity, allergenic activity, embriotoxicity or harmful effect on the reproductive function in animal and microbial models, or a cytotoxicity in human in vitro models were registered so far. Therefore it is concluded that alloferon has very low toxicity or is a practically nontoxic material.
15TABLE 15Summary of alloferon safety analysisActivityMethodResultsConclusionAcuteSingleMortality was not registered in any dose includingToxicometrictoxicitysubcutaneoushighest dose. No changes in animal growth, feeding,data areand intragastricmacroscopical structure of brain, inner and endocrinepositive.administration,organs as well as skin and subcutaneous cellularToxic/thera-500-6000 mg/kgtissue around injection place were found at the anypeutic dosein mice anddosage.ratio >35700300-5000 mg/kgtimes in ratsin ratsand >42800times in mice.Sub-0.2, 2 and 20None animal died during 90 day period.Alloferon hasacutemg/kg once aAnalysis of animal growth, feeding, rectalno toxicandday during 90temperature, ophthalmological (mucous surfaceseffects on thechronicdaysstate and eye morphometry), neuropsychologicalrats under longtoxicity(excitability threshold, spontaneous locomotorsterm dailyin ratsactivity) cardiovascular (systolic arterial pressure,administrationheart beating rate, ECG) liver (hexenal sleep),at therapeutickidney (urine composition, phenol red excretion),dose andhematological (hemogram, leukocyte formula,doses 10 andcoagulogram) indices, biochemical indices of100 timesperipheral blood (proteinemia, urea, creatinin,exceedingglucose, lipids, cholesterol, bilirubin, enzymatictherapeutic.activity, electrolyte composition), blood formingindices, pathomorphological and histological (heart,lungs, tracheas, stomach, pancreas, epithelialtissues, thymus, liver, spleen, kidney, adrenal, brain,testicles, ovary) data did not show negative changesin the organism's functions.Allerge-AnaphylacticNo signs of anaphylaxy were found at therapeuticAlloferon hasnicshockdosage. Minimal response (short term unrest, noseno detectableactivityscratch and quickened breath) was registered in 1allergenicinmale but 12 guinea pigs which obtained 10-foldactivity.guineatherapeutic dose.pigsImmuneThe reaction was not found under alloferoncomplexestherapeutic and 10-fold administrationreactionIndirect reactionFat cells degranulation rate was not changed underof fat cellsalloferon therapeutic and 10-fold administrationdegranulationConjunctiveThe reaction was not found under alloferonprobetherapeutic and 10-fold administrationReaction ofThe reaction was not found under alloferondelayedtherapeutic and 10-fold administrationhypersensitivityEmbrio-SubcutaneousAlloferon administration did not cause negativeAlloferon hastoxicityinjection 1.5 andeffects in the course of embryonic development andnoand15 mg/kg intopostnatal development in offsprings as well asembryotoxicinflu-pregnant ratsalterations of male and female reproductive functionactivity as wellence oninfluence onthetherepro-reproductiveductiveactivity in ratsfunctionMutage-Dominant lethalAlloferon in the dose 15 mg/kg did not induceAlloferon hasnicmutations in thedominant lethal mutations in the mouse germ cellsno mutagenicactivitymouse germand potentialcellscancerogenicactivityChromosomeAlloferon in the single dose 0.5 and 15 mg/kg andaberrations infivefold dose 0.5 mg/kg did not induce chromosomethe mouse boneaberrations in the mouse bone marrow cellsmarrow cellsAmes testAlloferon in the concentration range 0.1 - 1000microgram/Petri dish did not induce gene mutationsin 3 test line of Salmonella tiphimurium.DNA SOS-Alloferon has no DNA-damaging activity in the E. colireparationtest-line PQ37


[0101]


Claims
  • 1. Peptide consisting of up to 30 amino acid residues, characterized in that the peptide has the following general structural formula (1):
  • 2. Peptide of claim 1 consisting of up to 20 and preferably of 5-13 amino acid residues.
  • 3. Peptide of any of the preceding claims wherein X2 represents 0-3 amino acid residues, preferably one amino acid residue.
  • 4. Peptide of any of the preceding claims wherein XI is selected from the group consisting of nothing, His-Gly-Val-Ser-Gly-, Gly-Val-Ser-Gly-, Val-Ser-Gly-, Ser-Gly-, Pro-Ser-Leu-Thr-Gly-, Phe-lie-Val-Ser-Ala-, Thr-, Leu-Ala-Ser-Leu-, Cys-Val-Val-Thr-Gly-, Ile-Ser-Gly-, Cys-Gly-, Ile-Val-Ala-Arg-lle-, Phe-Gly-, His-Gly-Asp-Ser-Gly-, Ser-Gly- and Tyr-Ala-Met-Ser-Gly-.
  • 5. Peptide of any of the preceding claims wherein X2 is selected from the group consisting of a peptide bond, -Gin-, -Phe-, -Asp-, -Ser-, -Asn-, -Ala-, -Gln-Asn-, -Ala-Val- and -Ser-Asp-Gly-.
  • 6. Peptide of any of the preceding claims wherein X3 is selected from the group consisting of nothing -His-Gly, -His, -Tyr-Asp, -Phe-Val, -Pro, -GIn-His-Gly, -Leu-Ala, -Asp, -Pro-Leu, -Met and -Phe-Ile.
  • 7. Peptide of claim 1 which is selected from the group consisting of His-Gly-Val-Ser-Gly-His-Gly-Gln-His-Gly-Val-His-Gly, Gly-Val-Ser-Gly-His-Gly-Gln-His-Gly-Val-His-Gly, Val-Ser-Gly-His-Gly-Gln-His-Gly-Val-His, Ser-Gly-His-Gly-Gln-His-Gly-Val, Pro-Ser-Leu-Thr-Gly-His-Gly-Phe-His-Gly-Val-Tyr-Asp, Phe-Ile-Val-Ser-Ala-His-Gly-Asp-His-Gly-Val, Thr-His-Gly-Gln-His-Gly-Val, His-Gly-His-Gly-Val-His-Gly, Leu-Ala-Ser-Leu-His-Gly-Gln-His-Gly-Val, Cys-Val-Val-Thr-Gly-His-Gly-Ser-His-Gly-Val-Phe-Val, lle-Ser-Gly-His-Gly-Gln-His-Gly-Val-Pro, Cys-Gly-His-Gly-Asn-His-Gly-Val-His, lle-Val-Ala-Arg-lle-His-Gly-Gln-Asn-His-Gly-Val, His-Gly-Ser-Asp-Gly-His-Gly-Val-Gln-His-Gly, Phe-Gly-His-Gly-His-Gly-Val, His-Gly-Asn-His-Gly-Val-Leu-Ala, His-Gly-Asp-Ser-Gly-His-Gly-Gln-His-Gly-Val-Asp, His-Gly-His-Gly-Val-Pro-Leu, Ser-Gly-His-Gly-Ala-Val-His-Gly-Val-Met and Tyr-Ala-Met-Ser-Gly-His-Gly-His-Gly-Val-Phe-lle.
  • 8. Chemical compound exhibiting immunomodulatory activity comprising an amino acid sequence as defined in any of claims 1-7 or a pharmaceutically active salt or ether thereof, provided that the chemical compound is not a naturally occurring peptide or protein.
  • 9. Pharmaceutical composition comprising a peptide of any of claims 1-7 or a compound of claim 8 or a pharmaceutically active salt or ether of the peptide or compound.
  • 10. Use of a peptide of any of claims 1-7 or a compound of claim 8 or a pharmaceutically active salt or ether of the peptide or compound for the preparation of a pharmaceutical composition having immunomodulatory activity.
  • 11. Use of claim 10 wherein the pharmaceutical composition exhibits interferon inducing activity, antiviral activity, antitumor activity or stimulates the cytotoxic activity of human or animal lymphocytes.
  • 12. Use of claim 10 wherein the pharmaceutical composition is useful for the treatment or prophylaxis of immune deficient conditions, infections, such as viral infections or fungal systemic infections, or oncological diseases.
  • 13. Nucleotide sequence coding for a peptide of any of claims 1-7.
  • 14. Vector, suitable for the expression of a peptide according to any of claims 1-7 in a host cell, which expresses the peptide after transformation, comprising a DNA fragment coding for a peptide of any of claims 1-7.
  • 15. Host cell, characterized in that it is transformed by a vector according to claim 14.
  • 16. Host cell according to claim 15 characterized in that it is a bacterial cell.
Priority Claims (1)
Number Date Country Kind
99127725/04 Dec 1999 RU