WO1993006128A1 - Tnf antagonist peptides - Google Patents

Tnf antagonist peptides Download PDF

Info

Publication number
WO1993006128A1
WO1993006128A1 PCT/AU1992/000487 AU9200487W WO9306128A1 WO 1993006128 A1 WO1993006128 A1 WO 1993006128A1 AU 9200487 W AU9200487 W AU 9200487W WO 9306128 A1 WO9306128 A1 WO 9306128A1
Authority
WO
WIPO (PCT)
Prior art keywords
cys
gly
asp
pro
thr
Prior art date
Application number
PCT/AU1992/000487
Other languages
French (fr)
Inventor
Deborah Ann Rathjen
Fred Widmer
Original Assignee
Peptide Technology Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Peptide Technology Limited filed Critical Peptide Technology Limited
Publication of WO1993006128A1 publication Critical patent/WO1993006128A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to TNF antagonist peptides which have the ability to reduce TNF toxicity as manifest by decreased blood glucose levels and weight loss (cachexia).
  • the present invention further relates to compositions including these peptides as the active ingredient and methods of treatment involving the
  • LPS bacterial lipopolysaccharide
  • tumour necrosis factor alpha Associated with the injection of LPS is the extensive production of tumour necrosis factor alpha
  • TNF Tumor necrosis factor
  • Histopathology revealed severe leukostatsis in the lungs, haemorraghic necrosis in the adrenals, pancreas and other organs and tubular necrosis of the kidneys. All these changes were prevented if the animals were pretreated with a neutralizing monoclonal antibody against TNF.
  • TNF neutrophils by TNF.
  • TNF causes neutrophil degranulation, respiratory burst, enhanced antimicrobiocidal and
  • Endothelial cells are also an
  • TNF thrombomodulin
  • TNF a product of activated macrophages produced in response to infection and malignancy, was first identified as a serum factor in LPS treated mice which caused the haemorraghic necrosis of transplantable tumours in murine models and was cytoxoic for tumour cells in culture
  • Cachexia is a common symptom of
  • TNF also known as cachectin in the early literature
  • mice chronic administration of TNF (also known as cachectin in the early literature) to mice causes anorexia, weight loss and depletion of body lipid and protein within 7 to 10 days (Cerami et al., 1985 Immunol Lett 11, 173, Fong et al., 1989 J Exp Med 170, 1627).
  • TNF levels have been reported. These may be due to the short half-life of TNF (6 minutes), differences in TNF serum binding protein or true differences in TNF levels in chronic disease states.
  • TNF ⁇ as a mediator of inflammation
  • TNF has been measured in synovial fluid in patients with both rheumatoid and reactive arthritis and in the serum of patients with rheumatoid arthritis (Saxne et al., 1988 Arthrit. Rheumat. 31, 1041). Raised levels of TNF have been detected in renal transplant patients during acute rejection episodes (Maury and Teppo 1987 J. Exp Med 166, 1132). In animals TNF has been shown to be involved in the pathogenesis of graft versus host disease in skin and gut following allogeneic marrow transplantation.
  • TNF has also been shown to contribute significantly to the pathology of malaria (Clark et al., 1987; Am. J. Pathol. 129: 192-199). Further, elevated serum levels of TNF have been reported in malaria patients (Scuderi et al., 1986; Lancet 2: 1364-1365). TNF may also be shown to contribute significantly to the pathology of malaria (Clark et al., 1987; Am. J. Pathol. 129: 192-199). Further, elevated serum levels of TNF have been reported in malaria patients (Scuderi et al., 1986; Lancet 2: 1364-1365). TNF may also
  • TNF The biological response to TNF is mediated by
  • TNF-binding protein Infusion of TNF has been shown to cause an increase in circulating TNF-binding protein in humans (Lantz et al, 1990 Cytokine 2 1).
  • the present inventors have produced peptides which are able to reduce TNF toxicity as manifest by reduced mortality decreased blood glucose levels and weight loss (cachexia) in tumour-bearing mice treated with human recombinant TNF.
  • the present invention consists in a linear or cyclic peptide of the general formula:- x 1 -x 2 -x 3 -x 4 -x 5 -x 6 -x 7 -x 8 -x 9 -x 10 - in which x 1 is null, Cys or R 1 ;
  • x 2 is null, Cys, R 1 or
  • a 1 is Ser or Thr or Ala
  • a 2 is Lys or Arg or His
  • a 3 is Cys or Arg or His
  • a 4 is His or Lys or Arg or Phe or Tyr or Trp
  • a 5 is Lys or Arg or His
  • a 6 is Gly or Ala
  • a 7 is Thr or Ser or Ala
  • X 3 is Null, Cys, R 1 or A 8 -A 9
  • a 8 is absent or Gly or Ala or Tyr or Phe or
  • a 9 is Leu or lle or Val or Met
  • X 4 is Cys, R 1 or
  • a 11 is Asn or Gln
  • a 12 is Asp or Glu
  • a 13 is Cys or Arg or His
  • a 14 is Pro or Not - alkyl amino acid
  • X 5 is Gly or Ala
  • X 6 is Cys , R 2 or A 15 -A 16
  • a 15 is Pro or N ⁇ - alkyl amino acid
  • a 1 6 is
  • X 7 is null, Cys , R 2 or A 17 -A 18 -A 1 9 in which A 1 7 is Gln or Asn,
  • a 18 is Asp or Glu
  • a 19 is Thr or Ser or Ala
  • X 8 is null , Cys , R 2 , Asp, Glu, Gly or Ala,
  • X 9 is null , Cys , R 2 or
  • a 20 is Cys or Arg or His
  • a 21 is Arg or Lys or His
  • a 22 is Glu or Asp
  • a 23 is Cys or Arg or His
  • a 24 is Glu or Asp
  • a 25 is Ser or Thr or Ala
  • a 26 is Gly or Ala
  • a 27 is Ser or Thr or Ala
  • a 28 is Phe or Tyr or Trp or His
  • a 29 is Thr or Ser or Ala
  • X 10 is null, Cys or R 2
  • R 1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R 1 is glycosyl, nucleosyl, lipoyl or R 1 is an L- or D- ⁇ amino acid or an oligomer thereof consisting of up to 5 residues R 1 is absent when the amino acid adjacent is a desamino-derivative.
  • R 2 is -NR 12 R 13 , wherein R 12 and R 13 are
  • R 14 independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R 1 or N-glycosyl or N-lipoyl -OR 14 , where R 14 is H,
  • X 1 is always and only null when X 2 is R 1 , Cys or null
  • X 2 is always and only null when X 3 is R 1 , Cys or null
  • X 3 is always and only null when X 4 is R 1 or Cys
  • X 7 is always and only null when X 6 is R 2 or Cys
  • X 8 is always and only null when X 7 is null, R 2 or Cys
  • X 9 is always and only null when X 8 is null, R 2 or Cys
  • X 10 is always and only null when X 9 is null, R 2 or
  • a 17 -A 18 -A 19 and X 8 is Asp, Glu, Gly or Ala,
  • the peptide is selected from the group consisting of:-
  • the peptide is H-Leu-Tyr-Asn-Asp-Cys-Pro-Gly- Pro-Gly-Gln-Asp-Thr-Asp-OH.
  • the present invention consists in a linear or cyclic peptide of the general formula:-
  • Y 2 is Gly or Ala
  • Y 3 is Ala or Gla
  • Y 4 is Gln or Asn
  • Y 5 is Met or Gal or lle or Leu
  • Y 6 is Cys or Arg or His
  • Y 7 is Cys or Arg or His
  • Y 8 is Ser or Thr or Ala
  • Y 9 is Lys or Arg or His
  • Y 10 is Cys or Arg or His
  • Y 11 is Ser or Thr or Ala
  • Y 12 is Pro or n ⁇ - alkyl amino acid
  • Y 13 is Gly or Ala
  • Y 14 is Gln or Asn
  • Y 15 is His or Lys or Arg
  • Y 16 is Gly or Ala
  • Y 17 is null, Cys or A 2
  • R 1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R 1 is glycosyl, nucleosyl, lipoyl or R 1 is an L- or D- ⁇ amino acid or an oligomer thereof consisting of up to 5 residues R 1 is absent when the amino acid adjacent is a desamino-derivative.
  • R 2 is -NR 12 R 13 , wherein R 12 and R 13 are independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R 1 or N-glycosyl or N-lipoyl -OR 14 , where R 14 is H,
  • the peptide is Gly-Ala-Gln-Met-Cys(Acm)- Cys(Acm)-Ser-Lys-Cys(Acm)-Ser-Pro-mly-Gln-His-Gly.
  • the amino acids may be D or L isomers, however, generally the peptide will primarily consist of L-amino acids.
  • the cysteine residues may also include an Acn group protecting the cysteine residues.
  • the present invention consists in a pharmaceutical composition for use in treating subjects suffering from TNF toxicity, the composition comprising a peptide of the first or second aspect of the present invention in combination with a pharmaceutically
  • the present invention consists in a method of treating a subject suffering from the toxic effects of TNF, the method comprising administering to the subject the composition of the third aspect of the present invention.
  • the peptide is Peptide 371 as hereinafter defined.
  • the peptide of the present invention may be used in therapy to prevent TNF pathology associated with decreased blood glucose levels and weight loss and may be a useful therapy in the treatment of septic shock.
  • composition and method of the present invention would be expected to be useful as an
  • anti-inflammatory agent in a wide range of disease states including toxic shock, adult respiratory distress
  • meningitis cachexia, dermatitis, psoriasis, diabetes, neuropathy associated with infection or autoimmune
  • Guillame Barre Syndrome atherosclerosis, chronic fatigue syndrome, TB, other viral and parasitic diseases, OKT3 therapy, and would be expected to be useful in conjunction with radiation therapy, chemotherapy and transplantation, to ameliorate the toxic effects of such treatments or procedures.
  • composition and method of the present invention may also be useful in the treatment of diseases with an underlying element of local, systemic, acute or chronic inflammation.
  • composition and method of the present invention will be useful in treatment of any systemic or local infection leading to inflammation.
  • the peptides of the present invention may also be administered in cancer therapy in conjunction with
  • cytotoxic drugs which may potentiate the toxic effects of TNF ⁇ (Watanabe et al., 1988; Immunopharmacol.
  • Immunotoxicol. 10: 117-127) such as vinblastin, acyclovir, interferon alpha, cyclosporin A, IL-2, actinomycin D, adriamycin, mitomycin C, AZT, cytosine arabinoside, daunororubin, cis-platin, vincristine, 5-flurouracil and bleomycin; in cancer patients undergoing radiation
  • AIDS patients or others suffering from viral infection such as viral meningitis, hepatitis, herpes, green monkey virus etc.
  • immunostimulants such as thymopentin and muramyl peptides or cytokines such as IL-2 and GM-CSF.
  • peptides of the present invention will serve to abrogate the deleterious effects of TNF ⁇ .
  • non-conservative e.g., W-amino acids, desamino acids
  • G A; V, I, L, M; D, E; N, Q; S, T; K, R, H;
  • peptide is to be understood to embrace peptide bond replacements and/or peptide mimetics, i.e. pseudopeptides, as recognised in the art (see for example: Proceedings of the 20th European Peptide Symposium, edt. G. Jung. E. Bayer, pp. 289-336, and references therein), as well as salts and pharmaceutical preparations and/or formulations which render the bioactive peptide(s)
  • Such salts, formulations, amino acid replacements and pseudopeptide structures may be necessary and desirable to enhance the stability, formulation, deliverability (e.g., slow release, prodrugs), or to improve the economy of production, and they are
  • cyclic structure for stability such as N to C interchain imides and lactames (Ede et al in Smith and Rivier (Eds) "Peptides: Chemistry and Biology", Escom, Leiden (1991), p268-270), and sometimes also receptor binding may be enhanced by forming cyclic analogues.
  • An example of this is given in "Confirmationally restricted thymopentin-like compounds", U.S. pat. 4,457,489 (1985), Goldstein, G. et al.
  • ketomethylene, methylsulfide or retroinverse bonds to replace peptide bonds i.e. the interchange of the CO and NH moieties may both greatly enhance stability and
  • anti-idiotypic (anti-Ids) antibodies to an antigen to function like that antigen in its interaction with animal cells and components of cells.
  • anti-Ids anti-idiotypic antibodies
  • peptide hormone antigen can have hormone-like activity and interact specifically with the receptors to the hormone. Conversely, anti-Ids to a receptor can interact
  • anti-Ids bearing the internal image of an antigen can induce immunity to such an
  • anti-idiotypic antibodies are included within the scope of the present invention.
  • the present invention consists in an anti-idiotypic antibody to the peptide of the first aspect of the present invention, the
  • anti-idiotypic antibody being capable of reducing TNF toxicity.
  • CDRs Complementary Determining Regions
  • the present invention consists in a compound the three-dimensional structure of which is similar as a pharmacophore to the three- dimensional structure of the peptide of the first or second aspect of the present invention, the compound being characterized in that it reacts with antibodies raised against the peptide of the first aspect of the present invention and that the compound is capable of reducing TNF toxicity.
  • Figure 1 shows changes in blood glucose levels in ascites tumour-bearing mice in response to TNF treatment ⁇ 0 hrs, ⁇ 24 hrs;
  • Figure 2 shows weight loss by tumour-bearing mice as a result of TNF treatment
  • Figure 3 shows the effect of peptides on TNF-induced lethality, ⁇ 24 hrs post TNF; ⁇ 30 hrs post TNF.
  • Peptides numbered 363, 369-374 listed below were synthesized either by the Boc or Fmoc strategies.
  • Peptides were synthesized either on the continuous flow system using the standard Fmoc-polyamide method of solid phase peptide synthesis (Atherton et al, 1978, J. Chem. Soc. Chem. Commun., 13 , 537-539) or alternatively on the stirred cell system using polystyrene based resins.
  • the solid resin used was PepSyn KA which is a
  • the solid resin used was Fmoc-PepSyn K Am which is analogous polyamide resin with a Rink Linker, p-[(R,S)- ⁇ [1-(9H- fluoren-9-yl)-methoxyformamido]-2,4-dimethoxybenzyl]- phenoxyacetic acid (Bernatowicz et al, 1989, Tet. Lett. 30, 4645).
  • the synthesis starts by removing the Fmoc-group with an initial piperidine wash and
  • incorporation of the first amino acid is carried out by the usual peptide coupling procedure.
  • this is replaced by the Rink resin which is a polystyrene-based support.
  • Pentafluorophenyl active esters - the starting Fmoc amino acids are already in the active ester form.
  • Hydroxybenzotriazol esters are formed in situ either using Castro's reagent, BOP/NMM/HOBt (Fournier et al, 1989, Int. J. Peptide Protein Res., 33., 133-139) or using Knorr's reagent, HBTU/NMM/HOB-t (knorr et al, 1989, Tet. Lett., 10, 1927)
  • Peptides were cleaved from the PepSyn KA and PepSyn K Am using 5% water and 95% TFA where Arg(Pmc) is not present. Where Arg(Pmc) is present a mixture of 5% thioanisole in TFA is used. The cleavage typically took 3 h at room temperature with stirring. Thionanisole was removed by washing with ether or ethyl acetate and the peptide was extracted into an aqueous fraction. Up to 30% acetonitrile was used in some cases to aid dissolution. Lyophilization of the aqueous/acetonitrile extract gave the crude peptide.
  • Peptides from the Wang resin were cleaved in TFA containing 5% phenol, for up to 2 h at ambient temperature with stirring.
  • the peptide resin obtained after the synthesis was placed in a 0.3 M DMF solution of 10 equivalents of Ac-ONSu or acetic anhydride for 60 minutes.
  • the resin was filtered, washed with DMF, Ch 2 Cl 2 , ether and dried.
  • Peptides with C-terminal amides are synthesized on MBHA resins.
  • Acetylation was achieved by treating the deblocked resin with acetic anhydride in DMF solution.
  • Buffer system Buffer A - 0.1% aqueous TFA, Buffer B - 80% Acetonitrile and 20% A. Fractions are monitored by an analytical h.p.l.c. system with a diode array detection. Structures were confirmed by amino acid analysis, proton n.m.r. and FAB mass spectrometry.
  • the peptide resin (1.5 g) was treated with 15 ml HF at 0°C for 60 min. in the presence of p-cresol (1.5 g).
  • the peptide resin was washed with diethyl ether and then extracted with 30% aqueous acetonitrile. The peptide collected in the aqueous fraction was lyophilized to give
  • Buffer A 0.1% TFA/water and buffer B is 90% acetonitrile/water (0.1% TFA).
  • the gradient is 0-40% B over 60 min. (linear) monitored at 230 nm at a flow rate of 20m./min. A yield of 90 mg of 94% pure peptide was obtained.
  • Buffer A is 0.1% TFA and Buffer B is 80%
  • mice female, aged 10-12 weeks.
  • Figure 1 shows changes in blood glucose levels in ascites tumour-bearing mice inn response to TNF treatment. It can be seen that peptide 371 inhibited the TNF-induced
  • mice In lethality experiments Balb/C mice (female, aged 10 - 12 weeks) were primed intraperitoneally with pristane (0.5 ml) 10 days prior to peritoneal implantation of MethA tumour cells. Approximately 10 days later the mice were challenged with 25 ⁇ g recombinant human TNF administered subcutaneously and the number of mice surviving at 24 and 30 hours recorded. Mice were also treated with peptide 1 mg or anti-TNF monoclonal antibody (M47) by innoculation at a separate subcutaneous site. The results of these experiments are shown in Figure 3 and Table 3. TABLE 3
  • the peptides of the present invention are capable of preventing a decrease in blood glucose levels and weight loss in tumour-bearing mice treated with human recombinant tumour necrosis factor.
  • these peptides have utility in the treatment of numerous disease states which are due to the deleterious effects of TNF.

Abstract

The present invention provides TNF antagonist peptides which have the ability to reduce TNF toxicity. The present invention further relates to compositions including these peptides as the active ingredient and to methods of treatment involving the administration of this composition.

Description

TNF ANTAGONIST PEPTIDES
Field of the Invention
The present invention relates to TNF antagonist peptides which have the ability to reduce TNF toxicity as manifest by decreased blood glucose levels and weight loss (cachexia). The present invention further relates to compositions including these peptides as the active ingredient and methods of treatment involving the
administration of this composition.
Background of the invention
Many of the clinical features of Gram-negative septicemic shock may be reproduced in animals by the administration of bacterial lipopolysaccharide (LPS). The administration of LPS to animals can prompt severe
metabolic and physiological changes which can lead to death. Associated with the injection of LPS is the extensive production of tumour necrosis factor alpha
(TNF). Mice injected with recombinant human TNF develop piloerection of the hair (ruffling), diarrhea, a
withdrawn, unkempt appearance and die if sufficient amounts are given. Rats treated with TNF become
hypotensive, tachypneic and die of sudden respiratory arrest (Tracey et al., 1986 Science 234, 470). Severe acidosis, marked hemoconcentration and biphasic changes in blood glucose concentration were also observed.
Histopathology revealed severe leukostatsis in the lungs, haemorraghic necrosis in the adrenals, pancreas and other organs and tubular necrosis of the kidneys. All these changes were prevented if the animals were pretreated with a neutralizing monoclonal antibody against TNF.
The massive accumulation of neutrophils in the lungs of TNF-treated animals reflects the activation of
neutrophils by TNF. TNF causes neutrophil degranulation, respiratory burst, enhanced antimicrobiocidal and
anti-tumour activity (Klebanoff et al., 1986 J. Immunol. 136, 4220; Tsujimoto et al., 1986 Biochem Biophys Res Commun 137, 1094). Endothelial cells are also an
important target for the expression of TNF toxicity. TNF diminishes the anticoagulant potential of the endothelium, inducing procoagulant activity and down regulating the expression of thrombomodulin (Stern and Nawroth, 1986 J Exp Med 163, 740).
TNF, a product of activated macrophages produced in response to infection and malignancy, was first identified as a serum factor in LPS treated mice which caused the haemorraghic necrosis of transplantable tumours in murine models and was cytoxoic for tumour cells in culture
(Carswell et al., 1975 PNAS 72, 3666; Helson et al., 1975 Nature 258, 731). Cachexia is a common symptom of
advanced malignancy and severe infection. It is
characterised by abnormal lipid metabolism with
hypertriglyceridemia, abnormal protein and glucose
metabolism and body wasting. Chronic administration of TNF (also known as cachectin in the early literature) to mice causes anorexia, weight loss and depletion of body lipid and protein within 7 to 10 days (Cerami et al., 1985 Immunol Lett 11, 173, Fong et al., 1989 J Exp Med 170, 1627). These effects were reduced by concurrent
administration of antibodies against TNF, TNF has been measured in the serum of patients with cancer and chronic disease associated with cachexia. The results are
inconclusive since large differences in TNF levels have been reported. These may be due to the short half-life of TNF (6 minutes), differences in TNF serum binding protein or true differences in TNF levels in chronic disease states.
TNFα, as a mediator of inflammation, has been
implicated in the pathology of other diseases apart from toxic shock and cancer related cachexia. TNF has been measured in synovial fluid in patients with both rheumatoid and reactive arthritis and in the serum of patients with rheumatoid arthritis (Saxne et al., 1988 Arthrit. Rheumat. 31, 1041). Raised levels of TNF have been detected in renal transplant patients during acute rejection episodes (Maury and Teppo 1987 J. Exp Med 166, 1132). In animals TNF has been shown to be involved in the pathogenesis of graft versus host disease in skin and gut following allogeneic marrow transplantation.
Administration of a rabbit anti-murine TNF was
demonstrated to prevent the histological changes
associated with graft versus host disease and reduced mortality (Piquet et al., 1987 J Exp Med 166, 1280).
TNF has also been shown to contribute significantly to the pathology of malaria (Clark et al., 1987; Am. J. Pathol. 129: 192-199). Further, elevated serum levels of TNF have been reported in malaria patients (Scuderi et al., 1986; Lancet 2: 1364-1365). TNF may also
contribute to the brain pathology and consequent dementia observed in late stage HIV infections (Grimaldi et al Ann Nevrol 29 : 21)
The biological response to TNF is mediated by
specific cell surface receptors. At least two cell surface molecules of molecular weight 55 and 75 kd
specifically bind TNFα and TNF/3 with high affinity
(Hohmann et al, 1989 J. biol. Chem. 264 14927). These receptors have now been cloned (Loetscher et al, 1990 Cell 61, 351; Smith et al, 1990 Science 248 1019). Both TNF type I and II receptors are shed into serum following proteolytic cleavage to form serum binding proteins which reversibly inactivate TNF and which may regulate the activity of TNF in vivo (Kohno et al., 1990 PNAS 87 8331; Seckinger et al, 1988 J Biol Chem 264 11966). Infusion of TNF has been shown to cause an increase in circulating TNF-binding protein in humans (Lantz et al, 1990 Cytokine 2 1). The present inventors have produced peptides which are able to reduce TNF toxicity as manifest by reduced mortality decreased blood glucose levels and weight loss (cachexia) in tumour-bearing mice treated with human recombinant TNF.
Summary of the Invention
In a first aspect the present invention consists in a linear or cyclic peptide of the general formula:- x1-x2-x3-x4-x5-x6-x7-x8-x9-x10- in which x1 is null, Cys or R1;
x2 is null, Cys, R1 or
A1-A2-A3-A4-A5-A6-A7
in which A1 is Ser or Thr or Ala,
A2 is Lys or Arg or His,
A3 is Cys or Arg or His,
A4 is His or Lys or Arg or Phe or Tyr or Trp,
A5 is Lys or Arg or His,
A6 is Gly or Ala,
A7 is Thr or Ser or Ala,
X3 is Null, Cys, R1 or A8-A9
in which A8 is absent or Gly or Ala or Tyr or Phe or
Trp or His
A9 is Leu or lle or Val or Met
X4 is Cys, R1 or
Tyr or Phe or Trp or His or Gly or Ala,
A11 is Asn or Gln,
A12 is Asp or Glu,
A13 is Cys or Arg or His,
A14 is Pro or Not - alkyl amino acid
X5 is Gly or Ala,
X6 is Cys , R2 or A15-A16
in which A15 is Pro or Nα - alkyl amino acid, A 1 6 is
Gly or Ala,
X7 is null, Cys , R2 or A17-A18-A1 9 in which A1 7 is Gln or Asn,
A18 is Asp or Glu,
A19 is Thr or Ser or Ala,
X8 is null , Cys , R2 , Asp, Glu, Gly or Ala,
X9 is null , Cys , R2 or
A20-A21-A22-A23-A24-A25-A26-A27
-A 28-A29
in which A20 is Cys or Arg or His,
A21 is Arg or Lys or His,
A22 is Glu or Asp,
A23 is Cys or Arg or His,
A24 is Glu or Asp,
A25 is Ser or Thr or Ala,
A26 is Gly or Ala,
A27 is Ser or Thr or Ala,
A28 is Phe or Tyr or Trp or His,
A29 is Thr or Ser or Ala,
X 10 is null, Cys or R2
R1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R1 is glycosyl, nucleosyl, lipoyl or R1 is an L- or D-α amino acid or an oligomer thereof consisting of up to 5 residues R1 is absent when the amino acid adjacent is a desamino-derivative.
R2 is -NR12R13, wherein R12 and R13 are
independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R1 or N-glycosyl or N-lipoyl -OR14, where R14 is H,
straight, branched or cyclic alkyl, aralkyl or aryl, optionally substituted as defined for R1-O-glycosyl, -O-lipoyl or - an L- or D-α-amino acid or an oligomer thereof consisting of up to 5 residues or R2 is absent, when the adjacent amino acid is a decarboxy derivative of cysteine or a homologue thereof or the peptide is in a N-C cyclic form.
with the proviso that:
X1 is always and only null when X2 is R1, Cys or null
X2 is always and only null when X3 is R1, Cys or null
X3 is always and only null when X4 is R1 or Cys
X7 is always and only null when X6 is R2 or Cys
X8 is always and only null when X7 is null, R2 or Cys
X9 is always and only null when X8 is null, R2 or Cys
X10 is always and only null when X9 is null, R2 or
Cys
when X4 is R1 or Cys then X6 is A15-A16, X7 is
A17-A18-A19 and X8 is Asp, Glu, Gly or Ala,
when X6 is R2 or Cys then X4 is
A10-A11-A12-A13-A14
when X7 is null, R2 or Cys then X. is
A10-A11-A12-A13-A14.
In a preferred embodiment of the first aspect of the present invention the peptide is selected from the group consisting of:-
H-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-
Asp-OH,
Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gln-Asp-Thr-Asp-
Cys(Acm)-Arg-Glu-Cys(Acm)-Glu-Ser-Gly-Ser-Phe-Thr,
Thr-Lys-Cys(Acm)-His-Lys-Gly-Thr-Tyr-Leu-Tyr-Asn-Asp-
Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Asp,
Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly,
Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly,
Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly,
Gly-Pro-Gly-Gln-Asp-Thr-Asp,
Ac-Gly-Pro-Gly-Gln-Asp-Thr-Asp-NH2,
Gly-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Gly, and Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-NH2 In a particularly preferred embodiment of the present invention the peptide is H-Leu-Tyr-Asn-Asp-Cys-Pro-Gly- Pro-Gly-Gln-Asp-Thr-Asp-OH.
In a second aspect the present invention consists in a linear or cyclic peptide of the general formula:-
Y1-Y2-Y3-Y4-Y5-Y6-Y7-Y8-Y9-Y10- Y11-Y12-Y13-Y14-Y15-Y16-Y17
in which Y1 is null, Cys or R1
Y2 is Gly or Ala
Y3 is Ala or Gla
Y4 is Gln or Asn
Y5 is Met or Gal or lle or Leu
Y6 is Cys or Arg or His
Y7 is Cys or Arg or His
Y8 is Ser or Thr or Ala
Y9 is Lys or Arg or His
Y10 is Cys or Arg or His
Y11 is Ser or Thr or Ala
Y12 is Pro or nα - alkyl amino acid
Y13 is Gly or Ala
Y14 is Gln or Asn
Y15 is His or Lys or Arg
Y16 is Gly or Ala
Y17 is null, Cys or A2
R1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R1 is glycosyl, nucleosyl, lipoyl or R1 is an L- or D-α amino acid or an oligomer thereof consisting of up to 5 residues R1 is absent when the amino acid adjacent is a desamino-derivative.
R2 is -NR12R13, wherein R12 and R13 are independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R1 or N-glycosyl or N-lipoyl -OR14, where R14 is H,
straight, branched or cyclic alkyl, aralkyl or aryl, optionally substituted as defined for R.-O-glycosyl, -O-lipoyl or - an L- or D-α-amino acid or an oligomer thereof consisting of up to 5 residues or R2 is absent, when the adjacent amino acid is a decarboxy derivative of cysteine or a homologue thereof or the peptide is in a N-C cyclic form.
In a preferred embodiment of the second apect of the present invention the peptide is Gly-Ala-Gln-Met-Cys(Acm)- Cys(Acm)-Ser-Lys-Cys(Acm)-Ser-Pro-mly-Gln-His-Gly.
The amino acids may be D or L isomers, however, generally the peptide will primarily consist of L-amino acids. In addition, the cysteine residues may also include an Acn group protecting the cysteine residues.
In a third aspect the present invention consists in a pharmaceutical composition for use in treating subjects suffering from TNF toxicity, the composition comprising a peptide of the first or second aspect of the present invention in combination with a pharmaceutically
acceptable sterile carrier.
In a fourth aspect the present invention consists in a method of treating a subject suffering from the toxic effects of TNF, the method comprising administering to the subject the composition of the third aspect of the present invention.
In a preferred embodiment of the present invention the peptide is Peptide 371 as hereinafter defined.
The peptide of the present invention may be used in therapy to prevent TNF pathology associated with decreased blood glucose levels and weight loss and may be a useful therapy in the treatment of septic shock.
Further the composition and method of the present invention would be expected to be useful as an
anti-inflammatory agent in a wide range of disease states including toxic shock, adult respiratory distress
syndrome, hypersensitivity pneumonitis, systemic lupus erythromatosis, cystic fibrosis, asthma, bronchitis, drug withdrawal, schistosomiasis, sepsis, rheumatoid arthritis, acquired immuno-deficiency syndrome, multiple sclerosis, leperosy, malaria, systemic vasculitis, bacterial
meningitis, cachexia, dermatitis, psoriasis, diabetes, neuropathy associated with infection or autoimmune
disease, ischemia/reperfusion injury, encephalitis,
Guillame Barre Syndrome, atherosclerosis, chronic fatigue syndrome, TB, other viral and parasitic diseases, OKT3 therapy, and would be expected to be useful in conjunction with radiation therapy, chemotherapy and transplantation, to ameliorate the toxic effects of such treatments or procedures.
As the peptide of the present invention suppresses activation of neutrophils the composition and method of the present invention may also be useful in the treatment of diseases with an underlying element of local, systemic, acute or chronic inflammation. In general, it is believed the composition and method of the present invention will be useful in treatment of any systemic or local infection leading to inflammation.
The peptides of the present invention may also be administered in cancer therapy in conjunction with
cytotoxic drugs which may potentiate the toxic effects of TNFα (Watanabe et al., 1988; Immunopharmacol.
Immunotoxicol. 10: 117-127) such as vinblastin, acyclovir, interferon alpha, cyclosporin A, IL-2, actinomycin D, adriamycin, mitomycin C, AZT, cytosine arabinoside, daunororubin, cis-platin, vincristine, 5-flurouracil and bleomycin; in cancer patients undergoing radiation
therapy; and in AIDS patients (or others suffering from viral infection such as viral meningitis, hepatitis, herpes, green monkey virus etc.) and in patients receiving immunostimulants such as thymopentin and muramyl peptides or cytokines such as IL-2 and GM-CSF. In this use
peptides of the present invention will serve to abrogate the deleterious effects of TNFα .
It will be appreciated by those skilled in the art that a number of modifications may be made to the peptide of the present invention without deleteriously effecting the biological activity of the peptide. This may be achieved by various changes, such as insertions, deletions and substitutions (e.g., sulfation, phosphorylation, nitration, halogenation), either conservative or
non-conservative (e.g., W-amino acids, desamino acids) in the peptide sequence where such changes do not
substantially altering the overall biological activity of the peptide. By conservative substitutions the intended combinations are:-
G, A; V, I, L, M; D, E; N, Q; S, T; K, R, H;
F, Y, W, H; and P, Nα-alkylamino acids.
It may also be possible to add various groups to the peptide of the present invention to confer advantages such as increased potency or extended half-life in vivo, without substantially altering the overall biological activity of the peptide.
The term peptide is to be understood to embrace peptide bond replacements and/or peptide mimetics, i.e. pseudopeptides, as recognised in the art (see for example: Proceedings of the 20th European Peptide Symposium, edt. G. Jung. E. Bayer, pp. 289-336, and references therein), as well as salts and pharmaceutical preparations and/or formulations which render the bioactive peptide(s)
particularly suitable for oral, topical, nasal spray, ocular pulmonary, I.V., subcutaneous, as the case may be, delivery. Such salts, formulations, amino acid replacements and pseudopeptide structures may be necessary and desirable to enhance the stability, formulation, deliverability (e.g., slow release, prodrugs), or to improve the economy of production, and they are
acceptable, provided they do not negatively affect the required biological activity of the peptide.
Apart from substitutions, three particular forms of peptide mimetic and/or analogue structures of particular relevance when designating bioactive peptides, which have to bind to a receptor while risking the degradation by proteinases and peptidases in the blood, tissues and elsewhere, may be mentioned specifically, illustrated by the following examples: Firstly, the inversion of backbone chiral centres leading to D-amino acid residue structures may, particularly at the N-terminus, lead to enhanced stability for proteolytical degradation while not
impairing activity. An example is given in the paper "Tritriated D-ala1-Peptide T Binding", Smith, C.S. et al. Drug Development Res. 15, pp. 371-379 (1988).
Secondly, cyclic structure for stability, such as N to C interchain imides and lactames (Ede et al in Smith and Rivier (Eds) "Peptides: Chemistry and Biology", Escom, Leiden (1991), p268-270), and sometimes also receptor binding may be enhanced by forming cyclic analogues. An example of this is given in "Confirmationally restricted thymopentin-like compounds", U.S. pat. 4,457,489 (1985), Goldstein, G. et al. Finally, the introduction of
ketomethylene, methylsulfide or retroinverse bonds to replace peptide bonds, i.e. the interchange of the CO and NH moieties may both greatly enhance stability and
potency. An example of the latter type is given in the paper "Biologically active retroinverso analogues of thymopentin", Sisto A. et al in Rivier, J.E. and Marshall, G.R. (eds.) "Peptides, Chemistry, Structure and Biology", Escom, Leiden (1990), p.722-773. The peptides of the invention can be synthesized by various methods which are known in principle, namely by chemical coupling methods (cf. Wunsch, E.: "Methoden der organischen Chemie", Volume 15, Band 1 + 2, Synthese von Peptiden, Thieme Verlag, Stuttgart (1974), and Barrany, G.; Merrifield, R.B: "The Peptides", eds. E. Gross,
J. Meienhofer., Volume 2, Chapter 1, pp. 1-284, Academic Press (1980)), or by enzymatic coupling methods
(cf. Widmer, F., Johansen, J.T., Carlsberg Res. Commun., Volume 44, pp. 37-46 (1979), and Kullmann, W.: "Enzymatic Peptide Synthesis", CRC Press Inc., Boca Raton, Florida (1987), and Widmer, F., Johansen, J.T. in "Synthetic
Peptides in Biology and Medicine:, eds., Alitalo, K.,
Partanen, P., Vatieri, A., pp. 79-86, Elsevier, Amsterdam (1985)), or by a combination of chemical and enzymatic methods if this is advantageous for the process design and economy.
It will be seen that one of the alternatives embraced in the general formula set out above is for a cysteine residue to be positioned at both the amino and carboxy terminals of the peptide. This will enable the cylisation of the peptide by the formation of di-sulphide bond.
It is intended that such modifications to the peptide of the present invention which do not result in a decrease in biological activity are within the scope of the present invention.
As would be recognized by those skilled in the art there are numerous examples to illustrate the ability of anti-idiotypic (anti-Ids) antibodies to an antigen to function like that antigen in its interaction with animal cells and components of cells. Thus, anti-Ids to a
peptide hormone antigen can have hormone-like activity and interact specifically with the receptors to the hormone. Conversely, anti-Ids to a receptor can interact
specifically with a mediator in the same way as the receptor does. (For a review of these properties see: Gaulton, G.N. and Greane, M.I. 1986. Idiotypic mimicry of biological receptors, Ann. Rev. Immunol. 4, 253-280;
Sege, K and Peterson, P.A., 1978. Use of anti-idiotypic antibodies as cell surface receptor probes. Proc. Natl. Acad. Sci. U.S.A. 75, 2443-2447).
As might be expected from this functional similarity of anti-Id and antigen, anti-Ids bearing the internal image of an antigen can induce immunity to such an
antigen. (This nexus is reviewed in Hiernaux, J.R. 1988. Idiotypic vaccines and infectious diseases. Infect.
Immun. 56, 1407-1413.)
As will be appreciated by persons skilled in the art from the disclosure of this application it will be
possible to produce anti-idiotypic antibodies to the peptide of the present invention which will have similar biological activity. It is intended that such
anti-idiotypic antibodies are included within the scope of the present invention.
Accordingly, in a fifth aspect the present invention consists in an anti-idiotypic antibody to the peptide of the first aspect of the present invention, the
anti-idiotypic antibody being capable of reducing TNF toxicity.
The individual specificity of antibodies resides in the structures of the peptide loops making up the
Complementary Determining Regions (CDRs) of the variable domains of the antibodies. Since in general, the amino acid sequences of the CDR peptide loops of an anti-Id are not identical to or even similar to the amino acid
sequence of the peptide antigen from which it was
originally derived, it follows that peptides whose amino acid sequence is quite dissimilar, in certain contexts can take up a very similar three-dimensional structure. The concept of this type of peptide, termed a "functionally equivalent sequence" or mimotope by Geyson is familiar to those expert in the field. (Geyson, H.M. et al 1987.
Strategies for epitope analysis using peptide synthesis. J. Immun. Methods. 102, 259-274).
Moreover, the three-dimensional structure and function of the biologically active peptides can be simulated by other compounds, some not even peptidic in nature, but which mimic the activity of such peptides. This field of science is summarised in a review by
Goodman, M. (1990). (Synthesis, spectroscopy and computer simulations in peptide research. Proc. 11th American Peptide Symposium published in Peptides-Chemistry,
Structure and Biology pp 3-29. Ed Rivier, J.E. and
Marshall, G.R. Publisher ESCOM.)
As will be recognized by those skilled in the art, armed with the disclosure of this application, it will be possible to produce peptide and non-peptide compounds having the same three-dimensional structure as the peptide of the present invention. These "functionally equivalent structures" or "peptide mimics" will react with antibodies raised against the peptide of the present invention and may also be capable of reducing TNF toxicity. It is intended that such "peptide mimics" are included within the scope of the present invention.
Accordingly, in a sixth aspect the present invention consists in a compound the three-dimensional structure of which is similar as a pharmacophore to the three- dimensional structure of the peptide of the first or second aspect of the present invention, the compound being characterized in that it reacts with antibodies raised against the peptide of the first aspect of the present invention and that the compound is capable of reducing TNF toxicity.
More detail regarding pharmacophores can be found in Bolin et al. p 150, Polinsky et al. p 287, and Smith et al. p 485 in Smith and Rivier (Eds) "Peptides: Chemistry and Biology", Escom, Leiden (1991).
Detailed Description of the Invention
In order that the nature of the present invention may be more clearly understood, preferred forms thereof will now be described with reference to the following examples and Figures in which:-
Figure 1 shows changes in blood glucose levels in ascites tumour-bearing mice in response to TNF treatment □ 0 hrs, ■ 24 hrs; and
Figure 2 shows weight loss by tumour-bearing mice as a result of TNF treatment; and
Figure 3 shows the effect of peptides on TNF-induced lethality, ■ 24 hrs post TNF; ♡ 30 hrs post TNF.
Peptide Synthesis
Peptides numbered 363, 369-374 listed below were synthesized either by the Boc or Fmoc strategies.
(Abbreviations used in the following description: Acm
Acetamidomethyl, OCxl Cyclohexyl ester, Pmc
2,2,5,7,8-Pentamethylchroman-6-sulfonyl, NMM
N-Methylmorpholine).
363 H-Pro-Gln-Gly-Lys-Tyr-OH
369 H-Arg-Asp-Thr-Val-Cys(Acm)-Gly-Cys(Acm)-Arg-Lys-Asn-Gln
-Tyr-Arg-His-OH
370 Ac-Gln-Asp-Thr-Asp-Cys(Acm)-Arg-Glu-Cys(Acm)-Glu-Ser-
Gly-Ser-Phe-Thr-Ala-Ser-Glu-Asn-His-Leu-Arg-His-OH
371 H-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr -Asp-OH
372 H-Asp-Ser-Val-Cys(Acm)-Pro-Gln-Gly-Lys-Tyr-Ile-His-Pro- Gln-Asn-Asn-Ser-OH
373 H-Thr-Lys-Cys(Acm)-His-Lys-Gly-Thr-OH
374 H-Glu-Asn-Val-Lys-Gly-Thr-Glu-Asp-Ser-Gly-Thr-Thr-OH 466 Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gln
Asp-Thr-Asp-Cys(Acm)-Arg-Glu-Cys(Acm)-Glu-Ser-Gly-Ser- Phe-Thr 467 Thr-Lys-Cys(Acm)-His-Lys-Gly-Thr-Tyr-Leu-Tyr-Asn-Asp- Cys (Acm) -Pro-Gly-Pro-Gly-Gln-Asp-Thr-Asp
536 Arg-Glu-Asn-Glu-Cys(Acm)-Val-Ser-Cys(Acm)-Ser-Asn- Cys(Acm)-Thr-Lys-Leu
630 Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly
631 Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly
632 Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly
633 Gly-Pro-Gly-Gln-Asp-Thr-Asp
634 Ac-Gly-Pro-Gly-Gln-Asp-Thr-Asp-NH2
635 Gly-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Gly
636 Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr- NH2
637 Gly-Ala-Gln-Met-Cys(Acm)-Cys(Acm)-Ser-Lys-Cys(Acm)- Ser-Pro-Gly-Gln-His-Gly
SYNTHESIS OF PEPTIDES USING THE FMOC-STRATEGY
Peptides were synthesized either on the continuous flow system using the standard Fmoc-polyamide method of solid phase peptide synthesis (Atherton et al, 1978, J. Chem. Soc. Chem. Commun., 13 , 537-539) or alternatively on the stirred cell system using polystyrene based resins.
For peptides with free carboxyl at the C-terminus, the solid resin used was PepSyn KA which is a
polydimethylacrylamide gel on Kieselguhr support with 4-hydroxymethylphenoxyacetic acid as the functionalised linker (Atherton et al., 1975, J.Am.Chem.Soc. 97,
6584-6585). The carboxy terminal amino acid was attached to the solid support by a DCC/DMAP-mediated
symmetrical-anhydride esterification. The alternative is the Wang resin with Fmoc-amino acid previously attached.
For peptides with carboxamides at the C-terminus, the solid resin used was Fmoc-PepSyn K Am which is analogous polyamide resin with a Rink Linker, p-[(R,S)-α[1-(9H- fluoren-9-yl)-methoxyformamido]-2,4-dimethoxybenzyl]- phenoxyacetic acid (Bernatowicz et al, 1989, Tet. Lett. 30, 4645). The synthesis starts by removing the Fmoc-group with an initial piperidine wash and
incorporation of the first amino acid is carried out by the usual peptide coupling procedure. In the stirred cell system, this is replaced by the Rink resin which is a polystyrene-based support.
All Fmoc-groups during synthesis were removed by 20% piperidine/DMF and peptide bonds were formed either of the following methods except as indicated in Table 1:
1. Pentafluorophenyl active esters - the starting Fmoc amino acids are already in the active ester form.
2. Hydroxybenzotriazol esters - these are formed in situ either using Castro's reagent, BOP/NMM/HOBt (Fournier et al, 1989, Int. J. Peptide Protein Res., 33., 133-139) or using Knorr's reagent, HBTU/NMM/HOB-t (knorr et al, 1989, Tet. Lett., 10, 1927)
Side chain protection chosen for the amino acids was removed concomitantly during cleavage with the exception of Acm on cysteine which was left on after synthesis.
Figure imgf000019_0001
Cleavage Conditions
Peptides were cleaved from the PepSyn KA and PepSyn K Am using 5% water and 95% TFA where Arg(Pmc) is not present. Where Arg(Pmc) is present a mixture of 5% thioanisole in TFA is used. The cleavage typically took 3 h at room temperature with stirring. Thionanisole was removed by washing with ether or ethyl acetate and the peptide was extracted into an aqueous fraction. Up to 30% acetonitrile was used in some cases to aid dissolution. Lyophilization of the aqueous/acetonitrile extract gave the crude peptide.
Peptides from the Wang resin were cleaved in TFA containing 5% phenol, for up to 2 h at ambient temperature with stirring.
N-TERMINAL ACETYLATION
The peptide resin obtained after the synthesis (with Fmoc removed in the usual manner) was placed in a 0.3 M DMF solution of 10 equivalents of Ac-ONSu or acetic anhydride for 60 minutes. The resin was filtered, washed with DMF, Ch2Cl2, ether and dried.
SYNTHESIS OF PETIODES USING THE BOC-STRATEGY
Syntheses of these peptides were carried out using polystyrene based resins. For peptide with C-terminal acids, the appropriate Merrifield resin Boc-amino
acid-O-resin or the 100-200 mesh PAM resin is used.
Peptides with C-terminal amides are synthesized on MBHA resins.
Couplings of Boc-amino acids (Table 2) were carried out either using Symmetrical anhydride method or a HOBt ester method mediated by DCC or HTBU.
Figure imgf000021_0001
CLEAVAGE
Peptides were cleaved in HF with p-cresol or anisole as scavenger for up to 90 min. For His with Dnp
protection, the resin required pre-treatment with
mercaptoethanol:DIPEA:DMF (2:1:7), for 30 min. After removal of scavengers by ether wash, the crude peptide is extracted into 30% aqueous acetonitrile.
N-TERMINAL ACETYLATION
Acetylation was achieved by treating the deblocked resin with acetic anhydride in DMF solution.
PURIFICATION OF PEPTIDES
Crude peptide is purified by reverse phase
chromatography using either a C4 or C18 column and the
Buffer system: Buffer A - 0.1% aqueous TFA, Buffer B - 80% Acetonitrile and 20% A. Fractions are monitored by an analytical h.p.l.c. system with a diode array detection. Structures were confirmed by amino acid analysis, proton n.m.r. and FAB mass spectrometry.
Example 1
371 H-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-A sp-OH
The resin Boc-Asp (Cxl)-O-Resin (100-200 mesh 0.75 mM/g) was used in the synthesis. This is followed by the coupling of the subsequent residues in the following order:
Boc-Thr(Bzl)-OH DCC S.A. (Symmetrical anhydride)
Boc-Asp-(OCxl)-OH DCC S.A.
Boc-Gln-OH DCC/HOBt
Boc-Gly-OH DCC S.A.
Boc-Pro-OH DCC S.A.
Boc-Gly-OH DCC S.A.
Boc-Pro-OH DCC S.A.
Boc-Cys(Acm)-OH DCC/HOBt
Boc-Asp(OCxl)-OH DCC S.A.
Boc-Asn-OH DCC/HOBt
Boc-Tyr(2Br-Z) DCC S.A.
Boc-Leu-OH DCC S.A.
The peptide resin (1.5 g) was treated with 15 ml HF at 0°C for 60 min. in the presence of p-cresol (1.5 g).
The peptide resin was washed with diethyl ether and then extracted with 30% aqueous acetonitrile. The peptide collected in the aqueous fraction was lyophilized to give
570 mg.
Purification was carried out in a Waters Delta Prep instrument using a Delta Pak, C18 Prep-Pak cartridge, 300
A, 15 μm. Buffer A: 0.1% TFA/water and buffer B is 90% acetonitrile/water (0.1% TFA). The gradient is 0-40% B over 60 min. (linear) monitored at 230 nm at a flow rate of 20m./min. A yield of 90 mg of 94% pure peptide was obtained.
Amino acid analysis gave Asp 4.0 (4.0) Glu 1.1(1.0) Gly 2.0(2.0) Leu 1.0(1.0) Pro 1.9(2.0) Thr 1.0(1.0) Tyr
0.7(1.0) and Cys was present.
Fast atom bombardment mass spectroscopy using argon gas gave a M+H peak at 1465.
IV Example 2
374 H-Glu-Asn-Val-Lys-Gly-Thr-Glu-Asp-Ser-Gly-Thr-Thr-OH Pepsyn KA (2.0 g, 0.25 mMol/g) was esterified with
Fmoc-Thr-OH using 8 equiv of Fmoc-Thr(But) -OH, 4 equiv of
DCC and 0.4 equiv of DMAP.
The following amino acid derivatives and coupling conditions were used in subsequent steps:
Fmoc-Thr(But)-OH BOP/HOBt/NMM
Fmoc-Gly-OH BOP/HOBt/NMM
Fmoc-Ser(But)-OH BOP/HOBt/NMM
Fmoc-Asp(OBut)-OH BOP/HOBt/NMM
Fmoc-Glu(OBut)-OH BOP/HOBt/NMM
Fmoc-Thr(But)-OH BOP/HOBt/NMM
Fmoc-Gly-OH BOP/HOBt/NMM
Fmoc-Lys(Boc)-OH BOP/HOBt/NMM
Fmoc-Val-OH BOP/HOBt/NMM
Fmoc-Asn-OPfp
Fmoc-Glu(OBut)-OH BOP/HOBt/NMM
The peptide resin, after removal of the final Fmoc group weighed 2.76 g. Cleavage was carried in 95% aqueous
TFA and the crude peptide, after lyophilization gave
813 mg.
The crude peptide was purified on a Waters' Delta
Prep using a C18 Prep Pak cartridge 300 A 15 μm particle size. Buffer A is 0.1% TFA and Buffer B is 80%
Acetonitrile and 20% buffer A and a linear gradient of 0-80%B over 40 min monitored at 23 nm with a flow rate of
30 ml/min, gave 262 mg of the 90% pure peptide.
Amino acid analysis gave Asp 1.9(2.0) Glu 2.1(2.0)
Ser 0.9(1.0) Gly 2.3(2.0) Thr 2.9(3.0) Val 1.0(1.0) Lys
1.0(1.0). FAB mass spectrum of the peptide gives a M+H peak at 1237.
In Vivo Experiments
Balb/c mice (female, aged 10-12 weeks) were
inoculated with Meth A ascites tumour cells (2 x 10 ) two weeks before treatment with TNF (10μg) alone or in combination with TNF inhibiting peptide (10 mg, Fig. 1). At the time of treatment and 24 hrs after treatment
measurements were taken of weight and blood glucose levels. Figure 1 shows changes in blood glucose levels in ascites tumour-bearing mice inn response to TNF treatment. It can be seen that peptide 371 inhibited the TNF-induced
decrease in blood glucose levels which occurred within the 24 hours following TNF treatment. Peptide 371 also
inhibited weight loss by the tumour-bearing animals as a result of TNF treatment (Figure 2).
In lethality experiments Balb/C mice (female, aged 10 - 12 weeks) were primed intraperitoneally with pristane (0.5 ml) 10 days prior to peritoneal implantation of MethA tumour cells. Approximately 10 days later the mice were challenged with 25μg recombinant human TNF administered subcutaneously and the number of mice surviving at 24 and 30 hours recorded. Mice were also treated with peptide 1 mg or anti-TNF monoclonal antibody (M47) by innoculation at a separate subcutaneous site. The results of these experiments are shown in Figure 3 and Table 3. TABLE 3
EFFECT OF PEPTIDE 371 ON SURVIVAL OF MICE ADMINISTERED A LETHAL DOSE OF TNF
Figure imgf000024_0001
* 24 hours post TNF treatment As will be seen from the above results the peptides of the present invention, and in particular peptide 371, are capable of preventing a decrease in blood glucose levels and weight loss in tumour-bearing mice treated with human recombinant tumour necrosis factor. In the light of these results it is believed these peptides have utility in the treatment of numerous disease states which are due to the deleterious effects of TNF.
It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodimentswithout departing from the spirit or scope of the
invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.

Claims

CLAIMS :
1. A linear or cyclic peptide of the general formula:- x1-x2-x3-x4-x5-x6-x7-x8-x9-x10- in which x. is null, Cys or R1;
x2 is null, Cys, R1 or
A1-A2-A3-A4-A5-A6-A7
in which A1 is Ser or Thr or Ala,
A2 is Lys or Arg or His,
A3 is Cys or Arg or His,
A4 is His or Lys or Arg or Phe or Tyr or Trp,
A5 is Lys or Arg or His,
A6 is Gly or Ala,
A7 is Thr or Ser or Ala,
X3 is Null, Cys, R1 or A8-A9
in which A8 is absent or Gly or Ala or Tyr or Phe or Trp or His
A9 is Leu or lle or Val or Met
X4 is Cys, R1 or
A10-A11-A12-A13-A14 in Which A10 is
Tyr or Phe or Trp or His or Gly or Ala,
A11 is Asn or Gln,
A12 is Asp or Glu,
A13 is Cys or Arg or His,
A14 is Pro or Nα - alkyl amino acid
X5 is Gly or Ala,
X6 is Cys, R2 or A15-A16
in which A15 is Pro or Nα - alkyl amino acid, A 1 6 is
Gly or Ala,
X7 is null, Cys, R2 or A17-A18-A1 9
in which A17 is Gln or Asn,
A18 is Asp or Glu,
A19 is Thr or Ser or Ala,
X8 is null, Cys, R2, Asp, Glu, Gly or Ala, X9 is null, Cys, R2 or
A20-A21-A22-A23-A24-A25-A26-A27 -A 28-A29
in which A20 is Cys or Arg or His,
A21 is Arg or Lys or His,
A22 is Glu or Asp,
A23 is Cys or Arg or His,
A24 is Glu or Asp,
A25 is Ser or Thr or Ala,
A26 is Gly or Ala,
A27 is Ser or Thr or Ala,
A28 is Phe or Tyr or Trp or His,
A29 is Thr or Ser or Ala,
X 10 is null, Cys or R2
R1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R1 is glycosyl, nucleosyl, lipoyl or R1 is an L- or D-α amino acid or an oligomer thereof consisting of up to 5 residues R1 is absent when the amino acid adjacent is a- desamino-derivative.
R2 is -NR12R13, wherein R12 and R13 are
independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R1 or N-glycosyl or N-lipoyl -OR14, where R14 is H,
straight, branched or cyclic alkyl, aralkyl or aryl, optionally substituted as defined for R1-O-glycosyl, -O-lipoyl or - an L- or D-α-amino acid or an oligomer thereof consisting of up to 5 residues or R2 is absent, when the adjacent amino acid is a decarboxy derivative of cysteine or a homologue thereof or the peptide is in a N-C cyclic form,
with the proviso that:
X1 is always and only null when X2 is R1, Cys or null X2 is always and only null when X3 is R1, Cys or null X3 is always and only null when X4 is R1 or Cys
X7 is always and only null when X6 is R2 or Cys
X8 is always and only null when X7 is null, R2 or Cys X9 is always and only null when X8 is null, R2 or Cys X10 is always and only null when X9 is null, R2 or
Cys
when X4 is R1 or Cys then X6 is A15-A16, X7 is
A17-A18-A19 and X8 is Asp, Glu, Gly or Ala,
when X6 is R2 or Cys then X4 is
A10-A11-A12-A13-A14
when X7 is null, R2 or Cys then X4 is
A10-A11-A12-A13-A14.
2. A linear or cyclic peptide as claimed in claim 1 in which the peptide is selected from the group consisting of:-
H-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-
Asp-OH,
Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gln-Asp-Thr-Asp- Cys(Acm)-Arg-Glu-Cys(Acm)-Glu-Ser-Gly-Ser-Phe-Thr,
Thr-Lys-Cys(Acm)-His-Lys-Gly-Thr-Tyr-Leu-Tyr-Asn-Asp-
Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Asp,
Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly,
Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly,
Gly-Leu-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly,
Gly-Pro-Gly-Gln-Asp-Thr-Asp,
Ac-Gly-Pro-Gly-Gln-Asp-Thr-Asp-NH2,
Gly-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Gly, and
Ac-Tyr-Asn-Asp-Cys(Acm)-Pro-Gly-Pro-Gly-Gln-Asp-Thr-NH2
3. A linear or cyclic peptide as claimed in claim 1 in which the peptide is;
H-Leu-Tyr-Asn-Asp-Cys-Pro-Gly-Pro-Gly-Gln-Asp-Thr-Asp-OH.
4. A linear or cyclic peptide of the general formula:-
Y1-Y2-Y3-Y4-Y5-Y6-Y7-Y8-Y9-Y10- Y11-Y12-Y13-Y14-Y15-Y16-Y17 in which Y1 is null, Cys or R1
Y2 is Gly or Ala
Y3 is Ala or Gla
Y4 is Gln or Asn
Y5 is Met or Gal or lle or Leu
Y6 is Cys or Arg or His
Y7 is Cys or Arg or His
Y8 is Ser or Thr or Ala
Y9 is Lys or Arg or His
Y10 is Cys or Arg or His
Y11 is Ser or Thr or Ala
Y12 is Pro or nα - alkyl amino acid
Y13 is Gly or Ala
Y14 is Gln or Asn
Y15 is His or Lys or Arg
Y16 is Gly or Ala
Y17 is null, Cys or A2
R1 is R-CO, where R is H, straight, branched or cyclic alkyl up to C20, optionally containing double bonds and/or substituted with halogen, nitro, amino, hydroxy, sulfo, phospho or carboxyl groups (which may be substituted themselves), or aralkyl or aryl optionally substituted as listed for the alkyl and further including alkyl, or R1 is glycosyl, nucleosyl, lipoyl or R1 is an L- or D-α amino acid or an oligomer thereof consisting of up to 5 residues R1 is absent when the amino acid adjacent is a desamino-derivative.
R2 is -NR12R13, wherein R12 and R13 are
independently H, straight, branched or cyclic alkyl, aralkyl or aryl optionally substituted as defined for R1 or N-glycosyl or N-lipoyl -OR14, where R14 is H,
straight, branched or cyclic alkyl, aralkyl or aryl, optionally substituted as defined for R1-O-glycosyl, -O-lipoyl or - an L- or D-α-amino acid or an oligomer thereof consisting of up to 5 residues or R2 is absent. when the adjacent amino acid is a decarboxy derivative of cysteine or a homologue thereof or the peptide is in a N-C cyclic form.
5. A linear or cyclic peptide as claimed in claim 4 in which the peptide is;
Gly-Ala-Gln-Met-Cys(Acm)-Cys(Acm)-Ser-Lys-Cys(Acm)-Ser-Pro- Gly-Gln-His-Gly.
6. A pharmaceutical composition for use in treating subjects suffering from TNF toxicity, the composition comprising a peptide as claimed in any one of claims 1 to 5 in combination with a pharmaceutically acceptable
sterile carrier.
7. A method of treating a subject suffering from the toxic effects of TNF, the method comprising administering to the subject the composition as claimed in claim 6.
8. A method as claimed in claim 7 in which the subject is suffering from TNF pathology associated with decreased blood glucose levels and/or weight loss or from septic shock.
9. An anti-idiotypic antibody to the peptide as claimed in any one of claims 1 to 5, the anti-idiotypic antibody being characterised in that it is capable of reducing TNF toxicity.
10. A compound the three-dimensional structure of which is similar as a pharmacophore to the three- dimensional structure of the peptide as claimed in any one of claims 1 to 5, the compound being characterized in that it reacts with antibodies raised against the peptide as claimed in any one of claims 1 to 5 and that the compound is capable of reducing TNF toxicity.
PCT/AU1992/000487 1991-09-16 1992-09-16 Tnf antagonist peptides WO1993006128A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AUPK836791 1991-09-16
AUPK8367 1991-09-16

Publications (1)

Publication Number Publication Date
WO1993006128A1 true WO1993006128A1 (en) 1993-04-01

Family

ID=3775685

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU1992/000487 WO1993006128A1 (en) 1991-09-16 1992-09-16 Tnf antagonist peptides

Country Status (1)

Country Link
WO (1) WO1993006128A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486595A (en) * 1994-04-01 1996-01-23 Centecor, Inc. Tumor necrosis factor inhibitors
US5641751A (en) * 1995-05-01 1997-06-24 Centocor, Inc. Tumor necrosis factor inhibitors
US5753628A (en) * 1995-06-07 1998-05-19 Centocor, Inc. Peptide inhibitors of TNF containing predominantly D-amino acids
US6037324A (en) * 1996-01-04 2000-03-14 Leukosite, Inc. Inhibitors of MAdCAM-1-mediated interactions and methods of use therefor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003553A1 (en) * 1989-09-05 1991-03-21 Immunex Corporation TUMOR NECROSIS FACTOR-α AND -β RECEPTORS
GB2246569A (en) * 1990-06-15 1992-02-05 Charing Cross Sunley Research Tumour necrosis factor - alpha binding protein

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003553A1 (en) * 1989-09-05 1991-03-21 Immunex Corporation TUMOR NECROSIS FACTOR-α AND -β RECEPTORS
GB2246569A (en) * 1990-06-15 1992-02-05 Charing Cross Sunley Research Tumour necrosis factor - alpha binding protein

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CELL, Volume 61, issued April 1990, HANSRUEDI LOETSCHER et al., "Molecular Cloning and Expression of the Human 55kd Tumor Necrosis Factor Receptor", pages 351-359. *
CELL, Volume 61, issued April 1990, THOMAS J. SCHALL et al., "Molecular Cloning and Expression of a Receptor for Human Tumor Necrosis Factor", pages 361-370. *
DNA AND CELL BIOLOGY, Volume 9, No. 10, 1990, ADOLF HIMMLER et al., "Molecular Cloning and Expression of Human and Rat Tumor Necrosis Factor Receptor Chain (p60) and Its Soluble Derivative, Tumor Necrosis Factor-Binding Protein", pages 705-715. *
MOLECULAR AND CELL BIOLOGY, Volume 11, No. 6, issued June 1991, RAYMOND G. GOODWIN et al., "Molecular Cloning and Expression of the Type 1 and Type 2 Murine Receptors for Tumor Necrosis Factor", pages 3020-3026. *
PROC. NATL. ACAD. SCI. USA, Volume 87, issued November 1990, TADAHIKO KOHNO et al., "A Second Tumor Necrosis Factor Receptor Gene Product can Shed a Naturally Occuring Tumor Necrosis Factor Inhibitor", pages 8331-8335. *
PROC. NATL. ACAD. SCI. USA, Volume 87, issued October 1990, PATRICK W. GRAY et al., "Cloning of Human Tumor Necrosis Factor (TNF) Receptor cDNA and Expression of Recombinant Soluble TNF-Binding Protein", pages 7380-7384. *
SCIENCE, Volume 248, issued May 1990, CRAIG A. SMITH et al., "A Receptor for Tumor Necrosis Factor Defines an Unusual Family of Cellular and Viral Proteins", pages 1019-1023. *
THE EMBO JOURNAL, Volume 9, No. 10, 1990, YARON NOPHAR et al., "Soluble Forms of Tumor Necrosis Factor Receptors (TNF-Rs). The cDNA for the Type 1 TNF-R, Cloned Using Amino and Sequence Data of Its Soluble Form, Encodes Both the Cell Surface and a Soluble Form of the Receptor", pages 3269-3278. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486595A (en) * 1994-04-01 1996-01-23 Centecor, Inc. Tumor necrosis factor inhibitors
US5641751A (en) * 1995-05-01 1997-06-24 Centocor, Inc. Tumor necrosis factor inhibitors
US5753628A (en) * 1995-06-07 1998-05-19 Centocor, Inc. Peptide inhibitors of TNF containing predominantly D-amino acids
US6037324A (en) * 1996-01-04 2000-03-14 Leukosite, Inc. Inhibitors of MAdCAM-1-mediated interactions and methods of use therefor
US6274556B1 (en) 1996-01-04 2001-08-14 Millennium Pharmaceuticals, Inc. Inhibitors of MadCAM-1-mediated interactions and methods of use therefor

Similar Documents

Publication Publication Date Title
US5795859A (en) Peptide which abrogates TNF and/or LPS toxicity
CA1247602A (en) Rcrf and analogs
EP0275748B1 (en) Peptide derivatives and their therapeutical use
JPH03504013A (en) Peptide with T cell helper activity
JPH08511167A (en) Methods for enhancing the biological activity of chemokines
EP0298820A1 (en) Peptide derivatives and their therapeutic application
AU641366B2 (en) Peptide compounds
US5877276A (en) Polypeptide agonists for human interleukin-8
JPH02500517A (en) peptide compounds
HUT55803A (en) Process for producing vip-analogues ii
WO1996009062A1 (en) Polypeptide agonists and antagonists of human interleukin-8
JPH01501391A (en) Novel peptide
US5786335A (en) Sulfhydryl containing peptides for treating vascular disease
US5627156A (en) Polypeptide agonists for human interleukin-8
CA1243301A (en) Crf and analogs
WO1993006128A1 (en) Tnf antagonist peptides
US5587457A (en) Neutrophil stimulating peptides
JP2003500334A (en) RANTES-derived peptide having anti-HIV activity
US6375928B1 (en) Neutrophil stimulating peptides
US5912232A (en) Anti-inflammatory polypeptide antagonists of human I1-8
AU642487B2 (en) Neutrophil stimulating peptides
Zhao et al. Synthetic laminin-like peptides and pseudopeptides as potential antimetastatic agents
AU673332C (en) Peptide which abrogates TNF and/or LPS toxicity
JPH05505821A (en) CRF congeners
JP2010120937A (en) Inflammatory disease preventing and treating agent containing amino acid fragment of human apolipoprotein a-ii or its modification

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR CA CH CS DE DK ES FI GB HU JP KP KR LK LU MG MN MW NL NO PL RO RU SD SE US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL SE BF BJ CF CG CI CM GA GN ML MR SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA