WO2025193572A1 - Macrocyclic peptides useful as immunomodulators - Google Patents
Macrocyclic peptides useful as immunomodulatorsInfo
- Publication number
- WO2025193572A1 WO2025193572A1 PCT/US2025/019110 US2025019110W WO2025193572A1 WO 2025193572 A1 WO2025193572 A1 WO 2025193572A1 US 2025019110 W US2025019110 W US 2025019110W WO 2025193572 A1 WO2025193572 A1 WO 2025193572A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- followed
- fmoc
- phe
- acid
- lag
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/08—Linear peptides containing only normal peptide links having 12 to 20 amino acids
Definitions
- the present disclosure provides novel macrocyclic peptides which inhibit the LAG-3/MHC Class II protein/protein interaction, and are thus useful for the amelioration of various diseases, including cancer and infectious diseases.
- Lymphocyte activation gene-3 (LAG-3; LAG3; CD223) is a type I transmembrane protein that is expressed on the cell surface of activated CD4+ T cells, CD8+ T cells.
- T regulatory cells, B cells, and subsets of natural killer (NK) and dendritic cells (Triebel F, et al., J. Exp. Med. 1990; 171 : 1393-1405; Huard, Eur. J. Immunol. 1994; 24:3216-21; Grosso, J. Clin. Invest. 2007; 117:3383-92; Huang, Immunity. 2004; 21:503-13; Kieslow, Eur. J. Immunol.
- LAG-3 is closely related to CD4, which is a co-receptor for T helper cell activation. Both molecules have four extracellular Ig-Iike domains and require binding to their ligand, major histocompatibility complex (MHC) class II, for their functional activity'. In contrast to CD4, LAG-3 is only expressed on the cell surface of activated T cells and its cleavage from the cell surface terminates LAG-3 signaling. LAG-3 can also be found as a soluble protein but it does not bind to MHC class II and its function is unknown.
- MHC major histocompatibility complex
- LAG-3 is composed of the intracellular signalling domain, a transmembrane domain and 4 extracellular domains, designated DI to D4 (Huard 1997 Proc. Natl. Acad. Sci. 94:5744-9).
- Domain 1-2 associates with MHC class II ligand and it has been shown that the tip of domain 1 (extra loop) forms the binding site (Huard 1997 Proc. Natl. Acad. Sci. 94:5744-9).
- LAG-3 can also associate with alternative ligands, Galectin-3 and LSECtin. which induce its inhibitory signalling (Kouo 2015 Cancer Immunol Res. 3(4):412-23; Xu 2014 Cancer Res 74( 13): 3418-28). Association with Galectin-3 on cells or within the extracellular matrix could downregulate T cells that would not normally engage with MHC class II, such as CD8+ T cells. Therefore blockade of this ligand could serve as a mechanism for enhancing broad T cell function.
- LAG-3 A role of LAG-3 on T cells is to regulate T cell activation (Huard 1994 Eur. J. Immunol. 24:3216- 21). LAG-3 engages with MHC class II and this leads to down regulation of CD4+ T cells (Huard 1996 Eur. J. Immunol. 26:1180-6). Upon T cell activation, LAG-3 surface expression increases. The engagement of LAG-3 dimer with ligand induces signalling through an intracellular KIEELE domain (Workman 2002 J. Immunol 169:5392-5) leading to downregulation of the T cell activity. Therefore, LAG-3 serves to modulate responses to antigens, preventing over-stimulation and maintaining immune homeostasis.
- LAG-3 plays an important role in promoting regulatory T cell (Treg) activity and in negatively regulating T cell activation and proliferation (Workman CJ, et al., J. Tmmunok 2005; 174:688-695). Both natural and induced Treg express increased LAG-3, which is required for their maximal suppressive function (Camisaschi C. et al., J. Tmmunok 2010; 184:6545-6551 and Huang CT, et al, Immunity. 2004: 21 :503-513). Furthermore, ectopic expression of LAG-3 on CD4+ effector T cells reduced their proliferative capacity and conferred on them regulatory potential against third part) 7 T cells (Huang CT, et al, Immunity 7 .
- Epstein-Barr virus infection is yet another factor to consider in the potential induction of T cell exhaustion in hematological malignancies. It is known that EBVassociated CLL, Richter’s syndrome, and lymphoma cases are usually more aggressive than their EBV(-) counterpart (Tsimberidou AM, et al., Leuk Lymphoma 2006:47:827; Ansell SM, et al., Am J Hematol 1999;60:99.; Dolcetti R. et al., Infectious Agents and Cancer 2010;5:22; Kanakry JA, et al., Blood 2013;121:3547).
- LAG-3 expression has been evaluated as a prognostic or predictive marker in CLL and Hodgkin lymphoma (Zhang J, et al., BMC Bioinformatics 2010;! l(Suppl 9):S5; Kotaskova J, et al., J Mol Diagn 2010;12(3):328 — 334).
- LAG-3 expression on tumor-infiltrating lymphocytes (TILs) and peripheral blood also mediates T cell exhaustion in hematological malignancies (Dickinson JD, et al., Leuk Lymphoma 2006;47(2):231-44).
- LAG-3 blockade with specific antibodies has shown antitumor activity in leukemia (Berrien-Elliott. M, et al.. Cancer Research 2013; 73(2):605-616) and solid tumor models (Woo, S-R, et al., Cancer Research 2011; 72(4):917-927; Coding, S. R., et al., Journal of Immunology', Baltimore, Md. 1950; 190(9):4899-909). Therefore, LAG-3 is a potential therapeutic target in hematological malignancies.
- LAG-3 blockade with macrocyclic peptide inhibitors alone and in combination with standard of care (e.g., nivolumab. imatinib, lenalidomide) or with other checkpoint inhibitors deserves further exploration.
- standard of care e.g., nivolumab. imatinib, lenalidomide
- the molecules described herein demonstrate the ability to block the interaction of LAG-3 with MHC Class II, in both biochemical and cell-based experimental systems. These results are consistent with a potential for therapeutic administration to enhance immunity in cancer or chronic infection, including therapeutic vaccine.
- the macrocyclic peptides described herein are capable of inhibiting the interaction of Lag-3 with MHC class II. These compounds have demonstrated highly efficacious binding to LAG-3, blockade of the interaction of LAG-3 with MHC Class II, and are capable of promoting enhanced T cell functional activity, thus making them candidates for parenteral, oral, pulmonary, nasal, buccal and sustained release formulations.
- the macrocyclic peptides can possess one or more of the following functional properties described above, such as high affinity binding to human LAG-3, relatively good binding affinity to cyno LAG-3, and lack of binding to mouse LAG-3, the ability to inhibit binding of LAG-3 to MHC Class II molecules and/or the ability to stimulate antigen-specific T cell responses.
- R 16a is selected from hydrogen and Ci-Ce alkyl
- R 16 is selected from
- R 16 and R 16a together with the N atom attached formed a 4-6 cyclic rings, wherein the ring can be fused with another aromatic or heteroaromatic ring; the cyclic ring is subsituted with
- X’ is a chain of between 1 and 172 atoms wherein the atoms are selected from carbon and oxygen and wherein the chain may contain one, two, three, or four groups selected from -NHC(O)-, and -C(O)NH-, -NHC(O)NH-, embedded therein; and wherein the chain is optionally substituted with one to six groups independently selected from - CO2H, -C(O)NH2, and -(CH2)i-2CO2H, provided that X’ is other than unsubstituted PEG;
- R 30 is selected from -CO2H, -C(O)NR W R X , and -CH3 wherein R w and R x are independently selected from hydrogen and Ci-Cealkyl, tetrazole, and lithocolic acid and analogs of lithocolic acid;
- R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 . and R 12 are independently selected from a natural amino acid side chain and an unnatural amino acid side chain.
- the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof,
- R 16 is selected from
- R 16 and R 16a together with the N atom attached formed the sidechain of either D- or L-prohne, wherein the ring can be is substituted with
- a PEGn’ spacer can be inserted in any part of the R 16 (n’ is the number of -(CEECEEO)- unit in a PEG spacer); and wherein w’ is 1-5; m’ is 0-5; n' is 1-12;
- X’ is a chain of between 8 and 60 atoms wherein the atoms are selected from carbon and oxygen and wherein the chain may contain one, two, three, or four groups selected from -NHC(O)-, and -C(O)NH-, -NHC(O)NH-, embedded therein; and wherein the chain is optionally substituted with one to six groups independently selected from - CO2H, -C(O)NH2, and -(CH2)I-2CO2H, provided that X’ is other than unsubstituted PEG;
- R 30 is selected from -COzH, -C(O)NH 2 , and -CH3, tetrazole, and lithocolic acid and analogs of lithocolic acid;
- R 35 is selected from -CO 2 H, -C(O)NH 2 , CH3, tetrazole, and lithocolic acid and analogs; provided that at least one R 17 is other than hydrogen, -CEIs, or -CH2OH:
- the present disclosure provides a method of enhancing, stimulating, and/or increasing the immune response in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein.
- the method further comprises administering an additional agent prior to, after, or simultaneously with the macrocyclic peptide or peptides described herein.
- the additional agent is an antimicrobial agent, an antiviral agent, a cytotoxic agent, and/or an immune response modifier.
- the present disclosure provides a method of inhibiting growth, proliferation, or metastasis of cancer cells in a subject in need thereof, said method comprising administering to the subj ect a therapeutically effective amount of one or more macrocyclic peptides described herein.
- the cancer is selected from melanoma, renal cell carcinoma, squamous non-small cell lung cancer (NSCLC), non-squamous NSCLC, colorectal cancer, castration-resistant prostate cancer, ovarian cancer, gastric cancer, hepatocellular carcinoma, pancreatic carcinoma, squamous cell carcinoma of the head and neck, carcinomas of the esophagus, gastrointestinal tract and breast, and a hematological malignancy.
- NSCLC non-small cell lung cancer
- colorectal cancer colorectal cancer
- castration-resistant prostate cancer ovarian cancer
- gastric cancer hepatocellular carcinoma
- pancreatic carcinoma squamous cell carcinoma of the head and neck
- the present disclosure provides a method of treating an infectious disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein.
- the infectious disease is caused by a virus.
- the virus is selected from HIV, Hepatitis A, Hepatitis B, Hepatitis C, herpes virus, and influenza.
- the present disclosure provides a method of treating septic shock in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of one or more macrocyclic peptides described herein.
- the present disclosure provides a method blocking the interaction of LAG-3 with MHC Class II molecule in a subject, said method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein.
- R 1 group is selected from sidechain of the following amino acids: phenylalanine, ty rosine, try ptophan, leucine, 2-fluorophenylalanine, 3- fluorophenyl alanine, 4-fluorophenylalanine, 3,4-difluorophenylalanine. 3,5- difluorophenylalanine.
- 3-(4- thiazolyl)-alanine 4-acetamide-phenylalanine, homo-cyclohexylalanine, 2-fuanylalanine, homo-tyrosine, 4-phen oxy phenyl alanine, 4-cyclohexyloxyphenylalanme, 4- propargyl oxyphenyl alanine, ty rosine(O-aceti c aci d), 4-benoxyphenyl alanine, tiypophane(l -acetic acid), and 4-tert-butylphenylalanine;
- R 2 group is selected from sidechain of the following amino acids: alanine. 2-aminobutyric acid (Abu), cyclopropylglycine, threonine, and valine;
- R 3 group is selected from sidechain of the following amino acids: alanine, 3-(2-thienyl)-alanine, 3-(3-thienyl)-alanine, 3-benzothienylalanine (Bzt), 4-benzoylphenylalanine (Bpa), allothreonine, aspartic acid, citrulline (Cit).
- R 6 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 2-naphthylalanine (2-Nal), alanine, biphenylalanine (Bip), 3 ‘-carboxylic acid-biphenylalanine, 4’ -carboxylic acid- biphenylalanine, 3 -benzothienylalanine (Bzt), 4-benzoylphenylalanine (Bpa), phenylalanine, 3.4.5-trifluorophenylalanine.
- beta-hydroxy - phenylalanine isoleucine, histidine, leucine, phenyglycine (Phg), valine, tryptophan, tryptophan (1 -acetic acid), tryptophan (5-benzoxy), tyrosine, tyrosine (O-acetic acid), ty rosine (O-propargy 1), tyrosine (O-phenyl), and 2S)-2-amino-3-( ⁇ 4-[(2S)-2-amino-2- carboxy ethyl] phenyl ⁇ formamido)propanoic acid;
- R 8 group is selected from sidechain of the following amino acids: 4-cyclohexyloxyphenylalanine, 1-naphthylalanine (1-Nal), 2- naphthylalanine (2-Nal), (2S)-2-amino-3-[6-(2-methylphenyl)pyridin-3-yl]propanoic acid, biphenylalanine, 2'-methoxycarbonyl-biphenylalanine, 2'-ethyl-biphenylalanine, 2'- ethyl.
- R 9 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 6-quinolinylalanine, 8-isoquinolinylalanine, 4- isoquinolinylalanine, alanine (3-methoxy-isoquinolin-7-yl), 6-isoquinolinylalanine, 2- naphthylalanine (2-Nal), biphenylalanine, 4-benzoylphenylalanine (Bpa), 3- benzothienylalanine (Bzt), phenylalanine, 2-chlorophenylalanine.
- R 10 group is selected from sidechain of the following amino acids: aspartic acid, 4-carboxyphenylalanine, tyrosinetO-acetic acid), and try ptophanQ -acetic acid);
- R 11 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 2-naphthylalanine (2-Nal), 2-amino-3- (lH-indol-4-yl)propanoic acid, phenylalanine, 3,4-dimethoxyphenylalanine, 3- methoxyphenylalanine, 4-aminophenylalanine, 1 -methyltryptophane, asparagine, tryptophan, tyrosine, 5 -hydroxy tryptophane, and tyrosine(O-acetic acid); In compounds of formula (I).
- R 12 group is selected from sidechain of the following amino acids: alanine, alpha, alpha-dimethylglycine (Aib), aspartic acid, D- alanine, D-aspartic acid, D-glutamic acid, D-phenylalanine, D-histidine, D-homo- arginine, D-lysine, D-leucine, D-asparagine, D-glutamine, D-arginine, D-serine, D- threonine, D-v aline, D-try ptophan, D-tyrosine, glutamic acid, glycine, serine, tryptophan(l-scetivc acid). tyrosine(O-acetic acid), and tyrosine(O-propargyl);
- R 1 group is selected from sidechain of the following amino acids: L- Ala(P-3-pyridinyl) (L-3-Pya), L-Phe(4-COPh) (L-Bpa), D-Tyr(O-benzyl), L-Phe, L- Phe(2-F). L-Phe(2-Cl). L-Phe(3,4,5-tri-F).
- R 2 group is selected from sidechain of the following amino acids: L-Ala, L-Abu, L-Gly(a-cyclopropyl), and L-Val;
- R 3 group is selected from sidechain of the following amino acids: L-alloT, L-Asp, L-Phe(4-COOH), L-homo-Glu, L-Homo-Phe, L-homo-Gln, L-homo-Ser, L-Asn, L-Gln, L-Trp, L-Trp(l-CH 2 COOH). L-Trp(5-OBn), L-Tyr, and L-Tyr(CH 2 COOH);
- R 6 group is selected from sidechain of the following amino acids: L-l-Nal, L-2- Nal, L-Bip, L-Bip(3'-COOH), L-Bip(4'-COOH), L-Bpa, L-Bzt, L-Phe(3,4,5-tri-F), L- Phe(3,4-di-Cl), L-Phe(3,5-di-F), L-Phe(3-Cl), L-Phe(4-Cl), L-Phe(4-CN), L-Phe(4- CH 2 NH 2 ), L-Phe(4-CONH 2 ), L-Phe(4-COOH), L-Phe( Trp, L-Trp(l-CH 2 COOH), L-Trp(5-O-benzyl), L-Tyr, L-Tyr(CH 2 COOH), L-Tyr(phenyl), and L-Tyr(propargyl);
- R 8 group is selected from sidechain of the following amino acids: L- Tyr(cyclohexyl), L-2-Nal, L-Bip, L-Bip(2'-COOMe), L-Bip(2'-Et), L-Bip(2'-Et,4'-OH), L-Bip(2'-Et, ,4'-OMe), L-Bip(2'-F), L-Bip(2'-Me).
- R 9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Ala(P-quinolin-6-yl), L-Ala(P-isoquinolin-8-yl), L-Ala(P-isoquinolin-4-yl), L-Ala(P- quinolin-3-yl), L-Ala(P-3-methoxy-isoquinolin-7-yl), L-Bzt, L-Phe, L-Phe(2-Cl), L- Phe(3,4-di-F), L-Phe(3,5-di-F), L-Phe(3-CF 3 ), L-Phe(3-OMe), L-Phe(4-Cl), L-Phe(4- CONH2), L-homohomoPhe, L-homoSer(2-Me,4-Cl-phenyl), L-homoTyr, L-Trp, e), L-Trp(5-OH), L-T
- R 10 group is selected from sidechain of the following amino acids: L-Asp, L- Phe(4-COOH). and L-Tyr(CH 2 COOH);
- R 11 group is selected from sidechain of the following amino acids: L-l-Nal. L-2- Nal, L-Ala(P-indol-4-yl), L-Phe(3-OMe), L-Phe(4-NH 2 ), L-Trp, L-Trp(5-OH).
- R 12 group is selected from sidechain of the following amino acids: Aib,
- R 17 is each independently selected from hydrogen. -CH?, -(CH 2 ) Z NH 2 , - (C(R 17a ) 2 )o-4-X’-R 31 , -(CH 2 ) Z CO 2 H, -CH 2 OH, CH2C CH, and -(CH 2 )i- 3 -triazolyl-X’-R 35 , and R 35 is selected from -CO2H, -C(O)NH 2 , CH3, tetrazole, and lithocolic acid and analogs; provided that at least one R 17 is other than hydrogen, -CH?, or -CH 2 OH;
- R 1 group is selected from sidechain of the following amino acids: L-Phe(4-COPh) (L-Bpa), L-Phe, L-Phe(3,4,5-tri-F), L- L-Phe(3,5-di-F), L-Phe(3-CN), L-Phe(4-CN), L- Phe(4-CONH 2 ), L-Phe(4-COOH), L-Phe(3-CONH 2 ), L-Tyr(benzyl), and L- Tyr(CH 2 COOH):
- R 2 group is L-Val
- R 3 group is selected from sidechain of the following amino acids: L-alloT, L-Asp, L-Trp, L-Trp(l-CH 2 COOH), L-Trp(5-OBn), and L-Tyr;
- R’ group is selected from sidechain of the following amino acids: L-Asp, L-Dab, L-Dab(COCH 2 NH 2 ), D-His, D-Arg, Gly, L-Hyp, and L-Tyr(CH 2 COOH);
- R 6 group is selected from sidechain of the following amino acids: L-2-Nal, L- Bip(3'-COOH), L-Bip(4'-COOH), L-Phe(4-Cl), L-Phe(4-CH 2 NH 2 ), L-Phe(4-CONH 2 ), L- Phe(4-COOH), L-Trp, L-Trp(l-CH 2 COOH), L-Trp(5-O-benzyl), L-Tyr, L-Tyr(CH2C00H), L-Tyr(phenyl), and L-Tyr(propargy 1);
- R 7 group is selected from sidechain of the following amino acids: L-Dap, L- Phe(4-COOH). L-Asn. L-Ser, L-Thr, L-Tyr. and L-Tyr(propargyl);
- R 8 group is selected from sidechain of the following amino acids: L-Bip, L-Bip(2'- COOMe), L-Bip(2'-Et,4'-OH), L-Bip(2'-F), L-Bip(2'-Me), L-Bip(2-O-nBu), L-Bip(2'- OMe,4'-F), L-Bip(3',5'-di-F), L-Bip(3-CN), L-Bip(3'-COOH), L-Bip(3'-Me), L-Bip(4'- Cl).
- L-Phe(4-CH CH-Ph), and L- Tyr(Ph);
- R 9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Ala(P-quinolin-6-yl), L-Ala(P-isoquinolin-8-yl), L-Ala(P-isoquinolin-4-yl), L-Ala(P- quinolin-3-yl), L-Ala(P-3-methoxy-isoquinolin-7-yl), L-Bzt, L-Phe, L-Phe(2-Cl), L- Phe(3,4-di-F), L-Phe(3,5-di-F), L-Phe(3-CF 2 ), L-Phe(3-OMe), L-Phe(4-Cl), L-Phe(4- CONH 2 ), L-homohomoPhe, L-homoSer(2-Me,4-Cl-phenyl), L-homoTyr, L-Trp,
- R 10 group is L-Asp
- R 11 group is selected from sidechain of the following amino acids: L-Ala(P-indol- 4-yl), L-Trp, L-Trp(5-OH), and L-Tyr;
- R 12 group is selected from sidechain of the following amino acids:
- R 1 group is selected from sidechain of the following amino acids: L-Phe, L- Phe(3,5-di-F), L-Phe(4-CONH 2 ), L-Phe(4-COOH), and L-Phe(3-CONH 2 );
- R 3 group is selected from sidechain of the following amino acids: L-Asp. L-Trp, L-Trp(l-CH 2 COOH), and L-Tyr;
- R’ group is selected from sidechain of the following amino acids: L-Asp, D-Arg, and Gly;
- R 6 group is selected from sidechain of the following amino acids: L-Phe(4- COOH), L-Trp, and L-Tyr;
- R 7 group is L-Tyr
- R 8 group is selected from sidechain of the following amino acids: L-Bip, L-Bip(2'- Me), L-Bip(3'-COOH), L-Bip(4'-CONMe 2 ), L-Bip(4'-COOH), and L-Bip(4'-NHCOMe);
- R 9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Phe(3-CFs), L-Trp, and L-Tyr;
- R 10 group is L-Asp
- R 11 group is selected from sidechain of the following amino acids: L-Trp and L- Tyr;
- R 12 group is selected from sidechain of the following ammo acids: D-Ser. and Gly;
- an amino acid includes a compound represented by the general structure: where R and R' are as discussed herein.
- amino acid as employed herein, alone or as part of another group, includes, without limitation, an amino group and a carboxyl group linked to the same carbon, referred to as "a" carbon, where R and/or R' can be a natural or an un-natural side chain, including hydrogen.
- the absolute “S” configuration at the "a” carbon is commonly referred to as the “L” or “natural” configuration.
- the amino acid is glycine and is not chiral.
- naturally occurring amino acid side chain refers to side chain of any of the naturally occurring amino acids (i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, -histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, t rosine, and valine) usually in the S -configuration (i.e., the L-amino acid).
- naturally occurring amino acids i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, -histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, t rosine, and valine
- non-naturally occurring amino acid side chain refers to a side chain of any naturally occurring amino acid usually in the R-configuration (i.e.. the D-amino acid) or to a group other than a naturally occurring amino acid side chain in R- or S-configuration (i.e., the D- or L-amino acid, respectively) selected from:
- the "inhibitory concentration" of LAG-3 inhibitor is intended to mean the concentration at which a compound screened in an assay of the disclosure inhibits a measurable percentage of the interaction of LAG-3 with MHC Class II molecules.
- inhibitory concentration values range from IC50 to IC90, and are preferably, IC50, ICeo, IC70, ICso, or IC90, which represent 50%, 60%, 70%, 80% or 90% reduction in LAG-3/MHC Class II molecules binding activity, respectively. More preferably, the "inhibitory concentration” is measured as the IC50 value. It is understood that another designation for IC50 is the half-maximal inhibitor ⁇ ' concentration.
- Binding of the macrocyclic peptides to LAG-3 can be measured, for example, by methods such as homogeneous time-resolved fluorescence (HTRF), Surface Plasmon Resonance (SPR), isothermal titration calorimetry (ITC), nuclear magnetic resonance spectroscopy (NMR), and the like. Further, binding of the macrocyclic peptides to LAG- 3 expressed on the surface of cells can be measured as described herein in cellular binding assays.
- HTRF homogeneous time-resolved fluorescence
- SPR Surface Plasmon Resonance
- ITC isothermal titration calorimetry
- NMR nuclear magnetic resonance spectroscopy
- Administration of a therapeutic agent described herein includes, without limitation, administration of a therapeutically effective amount of therapeutic agent.
- therapeutically effective amount refers, without limitation, to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the LAG-3/MHC Class II molecules binding inhibitors described herein. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example and without limitation, treatment or prevention of the conditions listed herein.
- the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance.
- the disclosure pertains to methods of inhibiting growth of tumor cells in a subject using the macrocyclic peptides of the present disclosure.
- the macrocyclic peptides of the present disclosure are capable of binding to LAG-3, disrupting the interaction between LAG-3 and MHC class II molecules.
- the macrocyclic peptides of the present disclosure are potentially useful for modifying an immune response, treating diseases such as cancer or infectious disease, stimulating a protective autoimmune response or to stimulate antigen-specific immune responses (e.g., by coadministration of LAG-3 blocking peptides with an antigen of interest).
- Programmed Death Ligand 1 "Programmed Cell Death Ligand 1", “Protein PD-L1”. "PD-L1”, “PDL1”. "PDCDL1”, “hPD-Ll”, “hPD-LI”, “CD274" and “B7-H1” are used interchangeably, and include variants, isoforms, species homologs of human PD-L1, and analogs having at least one common epitope with PD-L1.
- the complete PD-L1 sequence can be found under GENBANK® Accession No. NP_054862.
- the complete PD-1 sequence can be found under GENBANK® Accession No. U64863.
- CTLA-4 cytotoxic T lymphocyte-associated antigen-4
- CTLA-4 antigen CD152
- CTLA-4 antigen CD152
- CTLA-4 nucleic acid sequence can be found under GENBANK® Accession No. L15006.
- immune response refers to the action of. for example, lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including macrocyclic peptides, cytokines, and complement) that results in selective damage to, destruction of, or elimination from the human body of invading pathogens, cells or tissues infected with pathogens, cancerous cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
- a “signal transduction pathway” refers to the biochemical relationship between a variety of signal transduction molecules that play a role in the transmission of a signal from one portion of a cell to another portion of a cell.
- cell surface receptor includes, for example, molecules and complexes of molecules capable of receiving a signal and the transmission of such a signal across the plasma membrane of a cell.
- An example of a “cell surface receptor” of the present disclosure is the PD-1 receptor.
- macrocyclic peptide derivatives refers to any modified form of the macrocyclic peptides disclosed herein, e.g., mutations, isofonns, peptides with altered linker backbones, conjugates with an antibody and/or another agent, etc..
- a (preferred?) macrocyclic peptide of the present disclosure that "specifically binds to human LAG-3" is intended to refer to a macrocyclic peptide that binds to human LAG-3 with an IC50 of less than about 1000 nM, less than about 300 nM, less than about less than about 100 nM, less than about 80 nM, less than about 60 nM, less than about 40 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 1 nM. or less.
- the term "about” shall be construed to mean anywhere between ⁇ 1, 2, 3, 4. 5, 6, 7. 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nM more or less than the cited amount.
- treatment refers to administering an active agent with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect a condition (e.g. , a disease), the symptoms of the condition, or to prevent or delay the onset of the symptoms, complications, biochemical indicia of a disease, or otherwise arrest or inhibit further development of the disease, condition, or disorder in a statistically significant manner.
- a condition e.g. , a disease
- any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
- a macrocyclic peptide of the disclosure comprises amino acid sequences that are homologous to the amino acid sequences of the macrocyclic peptides described herein, and wherein the macrocyclic peptides retain the desired functional and/or biological properties of the macrocyclic peptide of the disclosure.
- the disclosure provides a macrocyclic peptide, or antigen-binding portion thereof, comprising: an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the compounds described herein; and the macrocyclic peptide exhibits one or more of the following properties:
- the macrocyclic peptide binds to human LAG-3 with an IC50 of 200 nM or less;
- the macrocyclic peptide binds to human LAG-3 and one or more of the following: cynomolgus monkey LAG-3, and/or mouse LAG-3;
- the macrocyclic peptide inhibits tumor cell growth in a cellular assay and/or in vivo assay
- the macrocyclic peptide amino acid sequences may be about 80%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or about 99% homologous to the sequences set forth above.
- the term "about” shall be construed to mean anywhere between 1, 2, 3, 4, or 5 percent more or less than the cited amount.
- a macrocyclic peptide of the present disclosure having sequences with high identity (z.e., 80% or greater) to the sequences set forth above, can be obtained by mutating the sequences during chemical synthesis, for example, followed by testing of the altered macrocyclic peptide for retained function (i.e.. the functions set forth in (a) through (i) above) using the functional assays described herein.
- the biological and/or functional activity of the variant macrocyclic peptide amino acid sequences may be at least about lx, 2x, 3x, 4x, 5x, 6x,7x, 8x, 9x, or lOx more than the reference macrocyclic peptide on which the variant is based.
- the term "about” shall be construed to mean anywhere between O. lx, 0.2x, 0.3x, 0.4x, 0.5x, 0.6x, 0.7x, 0.8x, or 0.9x more or less than the cited amount.
- the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
- the percent identity between two amino acid sequences can be determined using the algorithm of Meyers E. et al., (Comput. Appl. Biosci., 4: 11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
- the percent identity between two amino acid sequences can be determined using the Needleman et al. (J. Mol. Biol., 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG® software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8. 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. Macrocyclic Peptides with Conservative Modifications
- a macrocyclic peptide of the disclosure comprises amino acid sequences that are homologous to the amino acid sequences of the macrocyclic peptides described herein, and wherein the macrocyclic peptides retain the desired functional and/or biological properties of the macrocyclic peptide of the disclosure.
- the disclosure provides a macrocyclic peptide, or antigen-binding portion thereof, comprising: an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the macrocyclic peptides described herein, wherein one or more amino acids have been substituted with a conservative amino acid; and the macrocyclic peptide exhibits one or more of the following properties:
- the macrocyclic peptide binds to human LAG-3 with an IC50 of 200 nM or less;
- the macrocyclic peptide binds to human LAG-3 and one or more of the following: cynomolgus monkey LAG-3, and/or mouse LAG-3;
- the macrocyclic peptide inhibits tumor cell growth in a cellular assay and/or in vivo assay
- conservative sequence modifications is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the macrocyclic peptide containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the disclosure by standard techniques know n in the art, such as substitution of peptide amidites during chemical synthesis, site- directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
- amino acids with basic side chains e.g., lysine, arginine, histidine
- acidic side chains e.g., aspartic acid, glutamic acid
- uncharged polar side chains e.g, glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
- nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
- beta-branched side chains e.g., threonine, valine, isoleucine
- aromatic side chains e.g.
- amino acid residues within the antigen binding regions of macrocyclic peptides of the disclosure can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i. e. , the functions set forth in (a) thru (i) above) using the functional assays described herein.
- Conservative amino acid substitutions may also be selected from one or more non-naturally occurring amino acids disclosed herein.
- the macrocyclic peptides of the present disclosure can be produced by methods known in the art, such as they can be synthesized chemically, recombinantly in a cell free system, recombinantly within a cell or can be isolated from a biological source. Chemical synthesis of a macrocyclic peptide of the present disclosure can be carried out using a variety of art recognized methods, including stepwise solid phase synthesis, semisynthesis through the conformationally-assisted re-ligation of peptide fragments, enzymatic ligation of cloned or synthetic peptide segments, and chemical ligation.
- a preferred method to synthesize the macrocyclic peptides and analogs thereof described herein is chemical synthesis using various solid-phase techniques such as those described in Chan, W.C. et al, eds., Fmoc Solid Phase Synthesis, Oxford University Press, Oxford (2000); Barany, G. et al, The Peptides: Analysis. Synthesis, Biology, Vol. 2 : "Special Methods in Peptide Synthesis, Part A", pp. 3-284, Gross, E. et al, eds., Academic Press, New York (1980); in Atherton, E., Sheppard, R. C. Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, Oxford, England (1989); and in Stewart, J. M.
- the peptides can be synthesized in a stepwise manner on an insoluble polymer support (also referred to as "resin") starting from the C-terminus of the peptide.
- a synthesis is begun by appending the C-terminal amino acid of the peptide to the resin through formation of an amide or ester linkage. This allows the eventual release of the resulting peptide as a C-terminal amide or carboxylic acid, respectively.
- the C-terminal amino acid and all other amino acids used in the synthesis are required to have their a-amino groups and side chain functionalities (if present) differentially protected such that the a-amino protecting group may be selectively removed during the synthesis.
- the coupling of an amino acid is performed by activation of its carboxyl group as an active ester and reaction thereof with the unblocked a-amino group of the N-terminal amino acid appended to the resin.
- the sequence of a-amino group deprotection and coupling is repeated until the entire peptide sequence is assembled.
- the peptide is then released from the resin with concomitant deprotection of the side chain functionalities, usually in the presence of appropriate scavengers to limit side reactions.
- the resulting peptide is finally purified by reverse phase HPLC.
- peptidyl-resins required as precursors to the final peptides utilizes commercially available cross-linked polystyrene polymer resins (Novabiochem, San Diego, CA; Applied Biosystems, Foster City, CA).
- Preferred solid supports are: 4- (2'.4'-dimethoxyphenyl-Fmoc-aminomethyl)-phenoxyacetyl-p-methyl benzhydrylamine resin (Rink amide MBHA resin); 9-Fmoc-amino-xanthen-3-yloxy-Merrifield resin (Sieber amide resin); 4-(9-Fmoc)aminomethyl-3,5- dimethoxyphenoxy)valerylaminomethyl-Merrifield resin (PAL resin), for C-terminal carboxamides.
- Coupling of first and subsequent amino acids can be accomplished using HOBt, 6-Cl-HOBt or HO At active esters produced from DIC/HOBt, HBTU/HOBt, BOP, PyBOP, or from DIC/6-Cl-HOBt, HCTU, DIC/HOAt or HATU, respectively.
- Preferred solid supports are: 2-chlorotrityl chloride resin and 9-Fmoc-amino-xanthen-3-yloxy- Merrifield resin (Sieber amide resin) for protected peptide fragments. Loading of the first amino acid onto the 2-chlorotrityl chloride resin is best achieved by reacting the Fmoc- protected amino acid with the resin in dichloromethane and DIEA. If necessary', a small amount of DMF may be added to solubilize the amino acid.
- the syntheses of the peptide analogs described herein can be carried out by using a single or multi-channel peptide synthesizer, such as an CEM Liberty Microwave synthesizer, or a Protein Technologies, Inc. Prelude (6 channels) or Symphony (12 channels) or Symphony X (24 channels) synthesizer.
- a single or multi-channel peptide synthesizer such as an CEM Liberty Microwave synthesizer, or a Protein Technologies, Inc. Prelude (6 channels) or Symphony (12 channels) or Symphony X (24 channels) synthesizer.
- the peptidyl-resin precursors for their respective peptides may be cleaved and deprotected using any standard procedure (see, for example, King, D.S. et aL Int. J. Peptide Protein Res., 36:255-266 (1990)).
- a desired method is the use of TFA in the presence of water, TIS as scavenger, and DTT or TCEP as the disulfide reducing agent.
- the peptidyl-resin is stirred in TFA/water/TIS/DTT (94:3:3: 1), v:v:v:w; 1 mL/100 mg of peptidyl resin) for 1.5-3 hrs at room temperature.
- the spent resin is then filtered off and the TFA solution was cooled and Et20 solution was added.
- the precipitates were collected by centrifuging and decanting the ether layer (3 x).
- the resulting crude peptide is either redissolved directly into DMF or DMSO or CH3CN/H2O for purification by preparative HPLC or used directly in the next step.
- Peptides with the desired purity can be obtained by purification using preparative HPLC, for example, on a Waters Model 4000 or a Shimadzu Model LC-8A liquid chromatography.
- the solution of crude peptide is injected into a YMC S5 ODS (20 x 100 mm) column and eluted with a linear gradient of MeCN in water, both buffered with 0.1% TFA, using a flow rate of 14-20 mL/min with effluent monitoring by UV absorbance at 220 nm.
- the structures of the purified peptides can be confirmed by electro-spray MS analysis.
- Mass Spectrometry “ESI-MS(+)” signifies electrospray ionization mass spectrometry performed in positive ion mode; “ESI-MS(-)” signifies electrospray ionization mass spectrometry’ performed in negative ion mode; L ‘ESI-HRMS(+)” signifies high-resolution electrospray ionization mass spectrometry performed in positive ion mode; ‘'ESI-HRMS(-)” signifies high-resolution electrospray ionization mass spectrometry’ performed in negative ion mode.
- the detected masses are reported following the “m/z' unit designation. Compounds with exact masses greater than 1000 were often detected as double-charged or triple-charged ions.
- the crude material was purified via preparative LC/MS. Fractions containing the desired product were combined and dried via centrifugal evaporation.
- Analytical LC/MS Condition C Column: Waters Acquity UPLC BEH Cl 8, 2.1 x 50 mm, 1.7-pm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate;
- Analytical LC/MS Condition D Column: Waters Acquity UPLC BEH C 18, 2.1 x 50 mm, 1.7-pm particles; Mobile Phase A: 5:95 acetonitrile:water with 0. 1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 70 °C; Gradient: 0-100% B over 3 minutes, then a 2.0-minute hold at 100% B; Flow: 0.75 mL/min; Detection: UV at 220 nm.
- Analytical LC/MS Condition E Column: Kinetex XB C18, 3.0 x 75 mm, 2.6-pm particles; Mobile Phase A: 10 mM ammonium formate in water: acetonitrile (98:2); Mobile Phase B: 10 mM ammonium formate in Water: acetonitrile (02:98); Gradient: 20- 100% B over 4 minutes, then a 0.6-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 254 nm.
- Analytical LC/MS Condition F Column: Ascentis Express C18, 2.1 x 50 mm, 2.7-pm particles; Mobile Phase A: 10 mM ammonium acetate in water: acetonitrile (95:5); Mobile Phase B: 10 mM ammonium acetate in Water: acetonitrile (05:95), Temperature: 50 °C; Gradient: 0-100% B over 3 minutes; Flow: 1.0 mL/min; Detection: UV at 220 nm.
- Analytical LC/MS Condition G Column: X Bridge C18, 4.6 x 50 mm, 5-qm particles; Mobile Phase A: 0.1% TFA in water; Mobile Phase B: acetonitrile, Temperature: 35 °C; Gradient: 5-95% B over 4 minutes; Flow: 4.0 mL/min; Detection: UV at 220 nm.
- BOP reagent benzotriazol- 1-yloxy-tris-dimethylamino-phosphonium hexafluorophosphate (Castro's reagent)
- PyBOP reagent benzotriazol-l-yloxy-tripyrrolidino phosphonium hexafluorophosphate
- HBTU 2-( I H-Benzotriazol- 1 -yl)- 1. 1.3.3-tetramethyluronim hexafluorophosphate
- HATU 0-(7-Azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronim hexafluorophosphate
- HCTU 2-(6-Chloro-l-H-benzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
- T3P 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide
- HOBT or HOBT*H2O 1 -hydroxy benzotriazole hydrate
- HO AT l-hydroxy-7-azabenzotriazole
- INT-1000 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ), and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
- the crude material was purified via preparative LC/MS with the following conditions: Column: Waters XBridge C18, 19 x 150 mm, 5 pm particles; Mobile Phase A: 5:95 acetonitrile: water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile: water with 0.1% trifluoroacetic acid; Gradient: 10-60% B over 20 minutes, then a 2-minute hold at 100% B; Flow: 40 mL/min.
- the yield of the product was 4.8 mg, and its estimated urity by LCMS analysis was 83.6%.
- INT-1002 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ) and the following procedures were done sequentially: “Tail loading procedure B” was followed with Fmoc-Lys(Boc)-OH, “Manual removal of Fmoc group procedure C” was followed, “Linker coupling procedure” was followed with Fmoc-12-amino-4,7,10-trioxadodecanoic acid, and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
- INT-1003 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol and the following procedures were done sequentially: “Tail loading procedure B” was followed with Fmoc-Lys(Boc)-OH, “Manual removal of Fmoc group procedure C” was followed, “Linker coupling procedure’' was followed with Fmoc-12-amino-4,7,10-trioxadodecanoic acid, and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
- the crude material was purified via preparative LC/MS.
- the yield of the product was 1.7 mg, and its estimated purity by LCMS analysis was 90%.
- INT-1004 to INT-1060 were prepared following similar procedures described for INT- 1000 to INT-1003 and general synthetic and purification procedures
- Example 1001 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ), and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
- Example 1002 was prepared as such: in a 25 mL glass solid support chemistry reaction vessel equipped with a frit were performed the following procedures sequentially:
- Linker coupling procedure ’ w as followed with l-(9-fluorenylmethyloxycarbonyl- amino)-4,7,10-trioxa-13-tridecanamine hydrochloride;
- Prelude Method C Double-coupling procedure w as followed with Fmoc-Gly-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Asp(OtBu)- OH;
- Example 1029 was prepared as follows; to a 25 mL polypropylene solid-phase reaction vessel was added 2-Chlorotrityl resin pre-loaded with (S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)pent-4-ynoic acid and the following procedures were done sequentially: '‘Tail loading procedure C” was followed with (S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)pent-4-ynoic acid;
- LAG-3 cell binding assay Human Raji cells expressing endogenous MHC Class II molecules were used for binding to either human LAG-3-mFc, mouse LAG-3, or cyno LAG-3-hFc proteins. Briefly Raji cells were plated in a 384-well plate (Coming 354663) at a density 7 of 8000 cells/well. After 2 hour incubation at a 37 °C and 5% CO2 incubator, LAG-3 antigen (hLAG-3 -mFc, mLAG-3-mFc, or cLAG-3-hFc) were added to all wells at a final concentration of 0.088. 0.25. or 0.072 pg/ml and incubated for 30 minutes.
- LAG-3 antigen hLAG-3 -mFc, mLAG-3-mFc, or cLAG-3-hFc
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biochemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
In accordance with the present disclosure, macrocyclic compounds have been discovered which inhibit the LAG-3/MHC Class II protein/protein interaction, and may be useful for the amelioration of various diseases, including cancer and infectious 5 diseases.
Description
MACROCYCLIC PEPTIDES USEFUL AS IMMUNOMODULATORS
CROSS REFERENCE
This application claims the benefit of U.S. Provisional Application No. 63/563,642 filed March 11, 2024 which is incorporated herein in its entirety'.
BACKGROUND
The present disclosure provides novel macrocyclic peptides which inhibit the LAG-3/MHC Class II protein/protein interaction, and are thus useful for the amelioration of various diseases, including cancer and infectious diseases.
Lymphocyte activation gene-3 (LAG-3; LAG3; CD223) is a type I transmembrane protein that is expressed on the cell surface of activated CD4+ T cells, CD8+ T cells. T regulatory cells, B cells, and subsets of natural killer (NK) and dendritic cells (Triebel F, et al., J. Exp. Med. 1990; 171 : 1393-1405; Huard, Eur. J. Immunol. 1994; 24:3216-21; Grosso, J. Clin. Invest. 2007; 117:3383-92; Huang, Immunity. 2004; 21:503-13; Kieslow, Eur. J. Immunol. 2005; 35:2081- 88: Workman CJ, et al., J. Immunol. 2009; 182(4): 1885- 91; Castelli, Oncoimmunology 2014; 3: 11). LAG-3 is closely related to CD4, which is a co-receptor for T helper cell activation. Both molecules have four extracellular Ig-Iike domains and require binding to their ligand, major histocompatibility complex (MHC) class II, for their functional activity'. In contrast to CD4, LAG-3 is only expressed on the cell surface of activated T cells and its cleavage from the cell surface terminates LAG-3 signaling. LAG-3 can also be found as a soluble protein but it does not bind to MHC class II and its function is unknown.
LAG-3 is composed of the intracellular signalling domain, a transmembrane domain and 4 extracellular domains, designated DI to D4 (Huard 1997 Proc. Natl. Acad. Sci. 94:5744-9). Domain 1-2 associates with MHC class II ligand and it has been shown that the tip of domain 1 (extra loop) forms the binding site (Huard 1997 Proc. Natl. Acad. Sci. 94:5744-9).
LAG-3 can also associate with alternative ligands, Galectin-3 and LSECtin. which induce its inhibitory signalling (Kouo 2015 Cancer Immunol Res. 3(4):412-23; Xu 2014 Cancer Res 74( 13): 3418-28). Association with Galectin-3 on cells or within the extracellular matrix could downregulate T cells that would not normally engage with
MHC class II, such as CD8+ T cells. Therefore blockade of this ligand could serve as a mechanism for enhancing broad T cell function.
A role of LAG-3 on T cells is to regulate T cell activation (Huard 1994 Eur. J. Immunol. 24:3216- 21). LAG-3 engages with MHC class II and this leads to down regulation of CD4+ T cells (Huard 1996 Eur. J. Immunol. 26:1180-6). Upon T cell activation, LAG-3 surface expression increases. The engagement of LAG-3 dimer with ligand induces signalling through an intracellular KIEELE domain (Workman 2002 J. Immunol 169:5392-5) leading to downregulation of the T cell activity. Therefore, LAG-3 serves to modulate responses to antigens, preventing over-stimulation and maintaining immune homeostasis.
It has been reported that LAG-3 plays an important role in promoting regulatory T cell (Treg) activity and in negatively regulating T cell activation and proliferation (Workman CJ, et al., J. Tmmunok 2005; 174:688-695). Both natural and induced Treg express increased LAG-3, which is required for their maximal suppressive function (Camisaschi C. et al., J. Tmmunok 2010; 184:6545-6551 and Huang CT, et al, Immunity. 2004: 21 :503-513). Furthermore, ectopic expression of LAG-3 on CD4+ effector T cells reduced their proliferative capacity and conferred on them regulatory potential against third part)7 T cells (Huang CT, et al, Immunity7. 2004; 21 :503-513). Recent studies have also shown that high LAG-3 expression on exhausted lymphocytic choriomeningitis virus (LCMV)-specific CD8+ T cells contributes to their unresponsive state and limits CD8+ T cell antitumor responses (Blackbum SD, et ak, Nat. Tmmunok 2009; 10:29-37 and Grosso JF, et ak, J. Clin. Invest. 2007; 117:3383-3392). In fact, LAG-3 maintained tolerance to self and tumor antigens via direct effects on CD8+T cells in 2 murine models (Grosso JF. et ak, J. Clin. Invest. 2007; 117:3383-3392).
Epstein-Barr virus infection is yet another factor to consider in the potential induction of T cell exhaustion in hematological malignancies. It is known that EBVassociated CLL, Richter’s syndrome, and lymphoma cases are usually more aggressive than their EBV(-) counterpart (Tsimberidou AM, et al., Leuk Lymphoma 2006:47:827; Ansell SM, et al., Am J Hematol 1999;60:99.; Dolcetti R. et al., Infectious Agents and Cancer 2010;5:22; Kanakry JA, et al., Blood 2013;121:3547). Interestingly, the expression of checkpoint inhibitors like PD-L1 and LAG-3 has also been documented in EBV-associated malignancies (Green MR, et al., Clin Cancer Res 2012; 18: 1611;
Monti S, et al., Blood 2005; 105: 1851). High expression of LAG-3 has in fact been documented in chronic viral infections and its blockade with anti-LAG-3 antibodies has been able to reduce viral titers and the expression of checkpoint inhibitors in murine models (Blackbum SD, et al., Nat. Immunol. 2009;10:29-37). Furthermore, LAG-3 expression, alone or in combination with other markers, has been evaluated as a prognostic or predictive marker in CLL and Hodgkin lymphoma (Zhang J, et al., BMC Bioinformatics 2010;! l(Suppl 9):S5; Kotaskova J, et al., J Mol Diagn 2010;12(3):328 — 334). LAG-3 expression on tumor-infiltrating lymphocytes (TILs) and peripheral blood also mediates T cell exhaustion in hematological malignancies (Dickinson JD, et al., Leuk Lymphoma 2006;47(2):231-44). Moreover, LAG-3 blockade with specific antibodies has shown antitumor activity in leukemia (Berrien-Elliott. M, et al.. Cancer Research 2013; 73(2):605-616) and solid tumor models (Woo, S-R, et al., Cancer Research 2011; 72(4):917-927; Coding, S. R., et al., Journal of Immunology', Baltimore, Md. 1950; 190(9):4899-909). Therefore, LAG-3 is a potential therapeutic target in hematological malignancies.
Recent preclinical studies have documented a role for LAG-3 in CD8 T cell exhaustion, and blockade of the LAG-3/ MHC Class II interaction using LAG-3 blocking antibodies or LAG-3-Ig fusion proteins is being evaluated in a number of clinical trials in cancer patients.
Additional background information can be found in WO2015/042246 Al, WP2015/116539 Al, and W02014/008218 AL
LAG-3 blockade with macrocyclic peptide inhibitors, alone and in combination with standard of care (e.g., nivolumab. imatinib, lenalidomide) or with other checkpoint inhibitors deserves further exploration.
The molecules described herein demonstrate the ability to block the interaction of LAG-3 with MHC Class II, in both biochemical and cell-based experimental systems. These results are consistent with a potential for therapeutic administration to enhance immunity in cancer or chronic infection, including therapeutic vaccine.
The macrocyclic peptides described herein are capable of inhibiting the interaction of Lag-3 with MHC class II. These compounds have demonstrated highly efficacious binding to LAG-3, blockade of the interaction of LAG-3 with MHC Class II, and are
capable of promoting enhanced T cell functional activity, thus making them candidates for parenteral, oral, pulmonary, nasal, buccal and sustained release formulations.
Additionally or alternatively, the macrocyclic peptides can possess one or more of the following functional properties described above, such as high affinity binding to human LAG-3, relatively good binding affinity to cyno LAG-3, and lack of binding to mouse LAG-3, the ability to inhibit binding of LAG-3 to MHC Class II molecules and/or the ability to stimulate antigen-specific T cell responses.
In its first embodiment the present disclosure provides a compound of formula (I)
(I), or a pharmaceutically acceptable salt thereof, wherein:
R16a is selected from hydrogen and Ci-Ce alkyl;
R16 is selected from
-(C(R17aR17))o-2-X’-R30, -(C(R17aR17))i-2C(O)N(R16a)C(R17a)2-X’-R31,
-C(R17aR17)i-2[C(O)N(R16a)C(R17aR17)i-2]w’ -X’-R31,
-(C(R17aR17)i-2C(O)NR16a)m -X’- R30;
-(C(R17aR17)i-2C(O)NR16a)m’-C(R17a)(R17)-CO2H;
-(C(R17aR17)i-2C(O)NR16a)m -C(R17a)(R17)-CO2H and
-(C(R17aR17))i-2-Phenyl-(C(R17aR17))i-2 -X’-R30; and wherein a PEGn- spacer can be inserted in any part of the R16 (n’ is the number of-(CH2CH2O)- unit in a PEG spacer);
Alternatively, R16 and R16a together with the N atom attached formed a 4-6 cyclic rings, wherein the ring can be fused with another aromatic or heteroaromatic ring; the cyclic ring is subsituted with
-(C(R17aR17))o-2-X’-R30,
-(C(R17aR17))i-2C(O)N(R16a)C(R17a)2-X’-R31.
-C(R17aR17)i-2[C(O)N(R16a)C(R17aR17)i-2PEGn’]w’ -X’-R31,
-(C(R17aR17)i-2C(O)NR16aPEGn )m -X’- R30; and
-(C(R17aR17)i-2C(O)NR16aPEGn’)m’-C(R17a)(R17)-CO2H;
-(C(R17aR17)i-2C(O)NR16a-PEGn )m’-C(R17a)(R17)-CO2H; and wherein a PEGn spacer can be inserted in any part of the R16 (n’ is the number of -(CH2CH2O)- unit in a PEG spacer); and wherein w’ is 1-5; m’ is 0-6; n’ is 1-20;
X’ is a chain of between 1 and 172 atoms wherein the atoms are selected from carbon and oxygen and wherein the chain may contain one, two, three, or four groups selected from -NHC(O)-, and -C(O)NH-, -NHC(O)NH-, embedded therein; and wherein the chain is optionally substituted with one to six groups independently selected from - CO2H, -C(O)NH2, and -(CH2)i-2CO2H, provided that X’ is other than unsubstituted PEG;
R30 is selected from -CO2H, -C(O)NRWRX, and -CH3 wherein Rw and Rx are independently selected from hydrogen and Ci-Cealkyl, tetrazole, and lithocolic acid and analogs of lithocolic acid;
R31 is -CO2H, -C(O)NRWRX, -CH3, alexa-5-SDP, tetrazole, and lithocolic acid and analogs of lithocolic acid, and biotin;
each R17a is independently selected from hydrogen, Ci-Cealkyl, -CH2OH, -(CH2)I- 4NH2, -(CH2)1-4NH-C(=NH)NH2, -CH2CO2H, -(CH2)2CO2H, each R17 is independently selected from hydrogen. -CH3, -(CH2)zNH2, - (C(R17a)2)o-4-X’-R31, -(CH2)ZCO2H, -CH2OH, CH2C5CH, and -(CH2)z-tnazolyl-X’-R35, wherein z is 1-6 and R35 is selected from -CO2H, -C(O)NRWRX, CH3, tetrazole, and lithocolic acid and analogs; provided that at least one R17 is other than hydrogen, -CH3, or -CH2OH;
R1, R2, R3, R5, R6, R7, R8, R9, R10, R11. and R12 are independently selected from a natural amino acid side chain and an unnatural amino acid side chain.
In a second aspect of the first embodiment: the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof,
When R,6a is selected from hydrogen, R16 is selected from
-(C(R17aR17))o-2-X'-R30,
-(C(R17aR17))i-2C(O)N(R16a)C(R17a)2-X'-R31.
-C(R17aR17)i-2[C(O)N(R16a)C(R17aR17)i-2]w- -X -R31.
-(C(R17aR17)i-2C(O)NR16a)m’-X - R30;
-(C(R17aR17)i-2C(O)NR16a)m -C(R17a)(R17)-CO2H;
-(C(R17aR17)i-2C(O)NR16a)m -C(R17a)(R17)-CO2H, and
-(C(R17aR17))i-2 -Phenyl-(C(R17aR17))i-2 -X’-R30; and wherein a PEGn spacer can be inserted in any part of the R16 (n’ is the number of-(CH2CH2O)- unit in a PEG spacer);
Alternatively, R16 and R16a together with the N atom attached formed the sidechain of either D- or L-prohne, wherein the ring can be is substituted with
-(C(R17aR17))o-2-X’-R30,
-(C(R17aR17))i-2C(O)N(R16a)C(R17a)2-X’-R31,
-C(R17aR17)i-2[C(O)N(R16a)C(R17aR17)i-2PEGn ]w -X’-R31.
-(C(R17aR17)i-2C(O)NR16aPEGn )m -X’- R30; and
-(C(R17aR17)i-2C(O)NR16aPEGn’)m -C(R17a)(R17)-CO2H;
-(C(R17aR17)i-2C(O)NR16a-PEGn’)m’-C(R17a)(R17)-CO2H; and wherein a PEGn’ spacer can be inserted in any part of the R16 (n’ is the number of -(CEECEEO)- unit in a PEG spacer); and wherein w’ is 1-5; m’ is 0-5; n' is 1-12;
X’ is a chain of between 8 and 60 atoms wherein the atoms are selected from carbon and oxygen and wherein the chain may contain one, two, three, or four groups selected from -NHC(O)-, and -C(O)NH-, -NHC(O)NH-, embedded therein; and wherein the chain is optionally substituted with one to six groups independently selected from - CO2H, -C(O)NH2, and -(CH2)I-2CO2H, provided that X’ is other than unsubstituted PEG;
R30 is selected from -COzH, -C(O)NH2, and -CH3, tetrazole, and lithocolic acid and analogs of lithocolic acid;
R31 is -CO2H, -C(O)NH2, -CH3, tetrazole, and hthocolic acid and analogs of lithocolic acid, and biotin; each R17a is independently selected from hydrogen, Ci-C2alkyl, -CEI2OH, -(CH2)i- 4NH2, -(CH2)1-4NH-C(=NH)NH2, -CH2CO2H, -(CH2)2CO2H, each R17 is independently selected from hydrogen, -CH3, -(CH2)zNH2, - (C(R17a)2)o-4-X’-R31, -(CH2)ZCO2H, -CH2OH, CH2C =CH. and -(CH2)i-3-triazolyl-X’-R35, and R35 is selected from -CO2H, -C(O)NH2, CH3, tetrazole, and lithocolic acid and analogs; provided that at least one R17 is other than hydrogen, -CEIs, or -CH2OH:
In another embodiment the present disclosure provides a method of enhancing, stimulating, and/or increasing the immune response in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein. In another embodiment the method
further comprises administering an additional agent prior to, after, or simultaneously with the macrocyclic peptide or peptides described herein. In another embodiment the additional agent is an antimicrobial agent, an antiviral agent, a cytotoxic agent, and/or an immune response modifier.
In another embodiment the present disclosure provides a method of inhibiting growth, proliferation, or metastasis of cancer cells in a subject in need thereof, said method comprising administering to the subj ect a therapeutically effective amount of one or more macrocyclic peptides described herein. In another embodiment the cancer is selected from melanoma, renal cell carcinoma, squamous non-small cell lung cancer (NSCLC), non-squamous NSCLC, colorectal cancer, castration-resistant prostate cancer, ovarian cancer, gastric cancer, hepatocellular carcinoma, pancreatic carcinoma, squamous cell carcinoma of the head and neck, carcinomas of the esophagus, gastrointestinal tract and breast, and a hematological malignancy.
In another embodiment the present disclosure provides a method of treating an infectious disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein. In another embodiment the infectious disease is caused by a virus. In another embodiment, the virus is selected from HIV, Hepatitis A, Hepatitis B, Hepatitis C, herpes virus, and influenza.
In another embodiment the present disclosure provides a method of treating septic shock in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of one or more macrocyclic peptides described herein.
In another embodiment the present disclosure provides a method blocking the interaction of LAG-3 with MHC Class II molecule in a subject, said method comprising administering to the subject a therapeutically effective amount of at least one macrocyclic peptide described herein.
In compounds of formula (I), R1 group is selected from sidechain of the following amino acids: phenylalanine, ty rosine, try ptophan, leucine, 2-fluorophenylalanine, 3- fluorophenyl alanine, 4-fluorophenylalanine, 3,4-difluorophenylalanine. 3,5- difluorophenylalanine. 3,4-dichlorophenyl, 3,4,5-difluorophenylalanine, pentafluorophenylalanine, 3-trifluoromethylphenylalanine, 4-methylphenylalanine, 4- chlorophenylalanine, 3-methoxyphenylalananie, 4-methoxyphenylalanine, 1-
naphthylalanine (1-Nal), 2-naphthyl alanine (2-Nal), 3-cyanophenylalanine, 4- cyanophenylalanine, 4-difluoromethylphenylalanine, biphenylalanine, 3-(2-thienyl)- alanine, 3-(3-thienyl)-alanine, 3-benzothienylalanine (Bzt), 4-bromophenylalanine, 3- pyridinylalanine, 4-trifluoromethylphenylalanine, 4-benzoylphenylalanine (Bpa), 3- chlorophenylalanine, 4-aminophenylalanine, 4-aminomethylphenylalanine, 4- carbamoylphenylalanine, 3-carbamoylphenylalanine, 4-carboxyphenylalaine. 3-(4- thiazolyl)-alanine, 4-acetamide-phenylalanine, homo-cyclohexylalanine, 2-fuanylalanine, homo-tyrosine, 4-phen oxy phenyl alanine, 4-cyclohexyloxyphenylalanme, 4- propargyl oxyphenyl alanine, ty rosine(O-aceti c aci d), 4-benoxyphenyl alanine, tiypophane(l -acetic acid), and 4-tert-butylphenylalanine;
In compounds of formula (I), R2 group is selected from sidechain of the following amino acids: alanine. 2-aminobutyric acid (Abu), cyclopropylglycine, threonine, and valine;
In compounds of formula (I), R3 group is selected from sidechain of the following amino acids: alanine, 3-(2-thienyl)-alanine, 3-(3-thienyl)-alanine, 3-benzothienylalanine (Bzt), 4-benzoylphenylalanine (Bpa), allothreonine, aspartic acid, citrulline (Cit). asparagine, glutamic acid, homo-glutamic acid, 2.3-diaminopropionic acid (Dap), phenylalanine, 2-fluorophenylalanine, 3-fluorophenylalanine, 4-fluorophenylalanine, 3- methylphenylalanine, 4-methylphenylalanine, 3-methoxyphenylalanine, 4- aminophenylalanine, 7-quinolinylalanine, homophenylalanine, homoglutamide, homeserine, leucine, 4-carboxyphenylalamine, tyrosine, glutamine, serine, 2-amino-4- aminobutyric acid (Dab), threonine, 4-carboxymethoxyphenyl-alanme, try ptophan, tryptophan (I-acetic acid), and tryptophne (5-benzoxy), tryptophan (1-methyl), tyrosine, and tyrosine(O-acetic acid).
In compounds of formula (I). R5 group is selected from sidechain of the following amino acids: alanine, citrulline, 1 -aminocyclopentane- 1 -carboxylic acid (Cle), 3-hydroxy 1-aminocyclopentane-l -carboxylic acid (Cle(3-OH)), aspartic acid, D-alanine, 2-amino- 4-aminobutyric acid (Dab), Dab(COCH2NH2), Dab(COCH2NHBoc), Dab(COCH2CH2NH2), Dab(COCH2CH2NHBoc), (S)-2-(X2-azaneyl)-4- guanidinobutanoic acid (Dab(-CH(=NH)NH2)), Dab(CO-ethyl), Dab(CH2-4- (CH2NH2)phenyl), Dab(CH2-4-(CH2NHBoc)phenyl), Dab(CONH-isopropyl), 2,3-
diaminopropionic acid (Dap), Dap(CH(=NH)NH2), D-glutamic acid, D-histidine, D- homoarginine, D-lyscine, D-lysine(COCH3), D-asparagine, D-glutamine. D-arginine, D- serine, D-valine, glutamine acid, phenylalanine, glycine, histidine, homoarginine, lyscine, leucine, asparagine, ornithine (Om), phenylglycine, propargylalanine, glutamine, arginine, valine, tryptophan (1 -acetic acid), tyrosine (O-acetic acid), ty rosine (O- propargyl), and p-alanine;
In compounds of formula (I). R6 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 2-naphthylalanine (2-Nal), alanine, biphenylalanine (Bip), 3 ‘-carboxylic acid-biphenylalanine, 4’ -carboxylic acid- biphenylalanine, 3 -benzothienylalanine (Bzt), 4-benzoylphenylalanine (Bpa), phenylalanine, 3.4.5-trifluorophenylalanine. 3,4-dichlorophenylalanine, 3,5- difluorophenylalanine, 3-chlorophenylalanine, 4-chlorophenylalanine, 4- cyanophenylalanine, 4-aminomethylphenylalanine, 4-carbamoylphenylalanine, 4- carboxyphenylalaine, 4-aminophenylalanine, alpha-methylphenylalanine. beta-hydroxy - phenylalanine, isoleucine, histidine, leucine, phenyglycine (Phg), valine, tryptophan, tryptophan (1 -acetic acid), tryptophan (5-benzoxy), tyrosine, tyrosine (O-acetic acid), ty rosine (O-propargy 1), tyrosine (O-phenyl), and 2S)-2-amino-3-({4-[(2S)-2-amino-2- carboxy ethyl] phenyl } formamido)propanoic acid;
In compounds of formula (I). R7 group is selected from sidechain of the following amino acids: alanine, aspartic acid, citrulline, 3-diaminopropionic acid (Dap), 2-amino-4- aminobutyric acid (Dab), Dap(COEt), Dap(CH(=NH)NH2), glutamic acid, glycine, histidine, 4-carboxyphenylalanine, homoarginine, lysine, asparagine, ornithine (Om), arginine, serine, threonine, tryptophan, try ptophan (1 -acetic acid), tyrosine, ty rosine (O- acetic acid), and tyrosine (O-propargyl);
In compounds of formula (I), R8 group is selected from sidechain of the following amino acids: 4-cyclohexyloxyphenylalanine, 1-naphthylalanine (1-Nal), 2- naphthylalanine (2-Nal), (2S)-2-amino-3-[6-(2-methylphenyl)pyridin-3-yl]propanoic acid, biphenylalanine, 2'-methoxycarbonyl-biphenylalanine, 2'-ethyl-biphenylalanine, 2'- ethyl. 4'-hydroxy-biphenylalanine, 2'-ethyl, 4'-methyoxy-biphenylalanine, 2'-fluoro- biphenylalanine, 2'-methyl-biphenylalanine. 2'-n-butoxy 1-biphenylalanine, 2'-methyoxy, 4'-fluoro-bipheny lalanine, 3', 5'-difluoro-bipheny lalanine, 3'-cyanobiphenylalanine, 3'-
carboxybiphenylalanine, 3'-methyl biphenylalanine, 4'-chlorobiphenylalanine, N- cyclopropyl-4'-carboxamide-biphenylalanine, 4'-dimethylcarbamoyl-biphenylalanine, 4'- carboxybiphenylalanine, 4'-acetamido-biphenylalanine, 4'-phenoxy -biphenylalanine, (3S)-3-amino-4-[5-(2-methylphenyl)pyrimidin-2-yl]-2-oxobutanoic acid, 4- benzoylphenylalanine (Bpa), (2S)-2-amino-3-[4-(l -benzofuran-2-yl)phenyl]propanoic acid, (2S)-2-amino-3-[4-(3-cyanothiophen-2-yl)phenyl]propanoic acid, (2S)-2-amino-3- {4-[(E)-2-phenylethenyl]phenyl}propanoic acid (Phe(4-CH=CH-phenyl)), 4- iodophenylalanine, 4-isopropylphenylalanine, 4-tert-butylphenylalanine, (2S)-2-amino-3- [4-(6-methoxypyridin-3-yl)phenyl]propanoic acid, (2S)-2-amino-3-[4-(pyrrolidine-l- carbonyl)phenyl]propanoic acid, homosenne(O-benzyl). tryptophan, tryptophan(5- benzoxy), tyrosine(O-propargyl), and tyrosine(O-phenyl);
In compounds of formula (I). R9 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 6-quinolinylalanine, 8-isoquinolinylalanine, 4- isoquinolinylalanine, alanine (3-methoxy-isoquinolin-7-yl), 6-isoquinolinylalanine, 2- naphthylalanine (2-Nal), biphenylalanine, 4-benzoylphenylalanine (Bpa), 3- benzothienylalanine (Bzt), phenylalanine, 2-chlorophenylalanine. 3,4- dichlorophenylalanine, 3,4-difluorophenylalanine, 3,5-difluorophenylalanine, 3- trifluoromethylphenylalanine, 3 -chlorophenylalanine, 3-methoxyphenylalanine, 4- chlorophenylalanine, 4-fluorophenylalanine, 4-aminophenylalanine, 4- carbamoylphenylalanine, 4-tert-butylphenylalanine, homohomophenylalanine, homoserine (O-2-methyl-4-chlorophenyl). homotyrosine, tryptophan, and tryptophan(l- acetic acid);
In compounds of formula (I), R10 group is selected from sidechain of the following amino acids: aspartic acid, 4-carboxyphenylalanine, tyrosinetO-acetic acid), and try ptophanQ -acetic acid);
In compounds of formula (I), R11 group is selected from sidechain of the following amino acids: 1 -naphthylalanine (1-Nal), 2-naphthylalanine (2-Nal), 2-amino-3- (lH-indol-4-yl)propanoic acid, phenylalanine, 3,4-dimethoxyphenylalanine, 3- methoxyphenylalanine, 4-aminophenylalanine, 1 -methyltryptophane, asparagine, tryptophan, tyrosine, 5 -hydroxy tryptophane, and tyrosine(O-acetic acid);
In compounds of formula (I). R12 group is selected from sidechain of the following amino acids: alanine, alpha, alpha-dimethylglycine (Aib), aspartic acid, D- alanine, D-aspartic acid, D-glutamic acid, D-phenylalanine, D-histidine, D-homo- arginine, D-lysine, D-leucine, D-asparagine, D-glutamine, D-arginine, D-serine, D- threonine, D-v aline, D-try ptophan, D-tyrosine, glutamic acid, glycine, serine, tryptophan(l-scetivc acid). tyrosine(O-acetic acid), and tyrosine(O-propargyl);
In another embodiment, compounds of formula (I), wherein, R1 group is selected from sidechain of the following amino acids: L- Ala(P-3-pyridinyl) (L-3-Pya), L-Phe(4-COPh) (L-Bpa), D-Tyr(O-benzyl), L-Phe, L- Phe(2-F). L-Phe(2-Cl). L-Phe(3,4,5-tri-F). L- L-Phe(3,5-di-F), L-Phe(3-Cl), L-Phe(3-CF3), L-Phe(3-CN), L-Phe(4-CN), L-Phe(4-CONH2), L-Phe(4-COOH), L- Phe(3-CONH2), L-Phe(penta-F), L-Tyr, L-Tyr(benzyl), L-Tyr(CH2COOH), L- Tyr(propargyl), and L-Tyr(phenyl);
R2 group is selected from sidechain of the following amino acids: L-Ala, L-Abu, L-Gly(a-cyclopropyl), and L-Val;
R3 group is selected from sidechain of the following amino acids: L-alloT, L-Asp, L-Phe(4-COOH), L-homo-Glu, L-Homo-Phe, L-homo-Gln, L-homo-Ser, L-Asn, L-Gln, L-Trp, L-Trp(l-CH2COOH). L-Trp(5-OBn), L-Tyr, and L-Tyr(CH2COOH);
R3 group is selected from sidechain of the following amino acids: L-Dap(- C(=NH)NH2), L-Cit. L-Asp. L-Dab. L-Dab(CH2Phenyl-4-CH2NH2), L- Dab(COCH2CH2NH2), L-Dab(COCH2NH2), L-Dab(COEt), Dab(G), L-Dap, D-Glu, D- His, D-Asn, D-Gln, D-Arg, D-Val, L-Glu, L-Phe, Gly, L-His, L-Hyp, L- Dab(COCH2NHBoc), L-Dab(COCH2CH2NHBoc), L-Asn, L-Om, L-Pro, L-Pro(NH- C(=NH)NH2), L-Pro(4-NH2), L-Phg, L-Pra, L-Gln, L-Arg, L-Val, L-Trp(l-CH2COOH), L-Tyr(CH2COOH), and L-Tyr(Pra),
R6 group is selected from sidechain of the following amino acids: L-l-Nal, L-2- Nal, L-Bip, L-Bip(3'-COOH), L-Bip(4'-COOH), L-Bpa, L-Bzt, L-Phe(3,4,5-tri-F), L- Phe(3,4-di-Cl), L-Phe(3,5-di-F), L-Phe(3-Cl), L-Phe(4-Cl), L-Phe(4-CN), L-Phe(4-
CH2NH2), L-Phe(4-CONH2), L-Phe(4-COOH), L-Phe(
Trp, L-Trp(l-CH2COOH), L-Trp(5-O-benzyl), L-Tyr, L-Tyr(CH2COOH), L-Tyr(phenyl), and L-Tyr(propargyl);
R7 group is selected from sidechain of the following amino acids: L- Dap(C(=NH)NH2). L-Dab. L-Dap, L-Phe(4-COOH), Gly. L-His, L-homoArg, L-Lys, L- Asn, L-Om, L-Arg, L-Ser, L-Thr, L-Trp, L-Tyr, L-Tyr(CH2COOH), L-Tyr(propargyl), and L-Trp(CH2COOH);
R8 group is selected from sidechain of the following amino acids: L- Tyr(cyclohexyl), L-2-Nal, L-Bip, L-Bip(2'-COOMe), L-Bip(2'-Et), L-Bip(2'-Et,4'-OH), L-Bip(2'-Et, ,4'-OMe), L-Bip(2'-F), L-Bip(2'-Me). L-Bip(2-O-nBu), L-Bip(2'-OMe,4'-F), L-Bip(3',5'-di-F), L-Bip(3-CN), L-Bip(3'-COOH), L-Bip(3'-Me), L-Bip(4'-Cl), L-Bip(4'- CONH-cyclopropyl), L-Bip(4'-CONMe2), L-Bip(4'-COOH), L-Bip(4'-NHCOMe), L- Bip(4'-OPh), L-Bpa, L-Phe(4-(2 -benzofuranyl)), L-Phe(4-(2-(3-CN-thienyl)), L-Phe(4- CH=CH-Ph). L-Phe(4-iodo). (2S)-2-amino-3-[4-(6-methoxypyridin-3- yl)phenyl]propanoic acid, and L-Tyr(Ph);
R9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Ala(P-quinolin-6-yl), L-Ala(P-isoquinolin-8-yl), L-Ala(P-isoquinolin-4-yl), L-Ala(P- quinolin-3-yl), L-Ala(P-3-methoxy-isoquinolin-7-yl), L-Bzt, L-Phe, L-Phe(2-Cl), L- Phe(3,4-di-F), L-Phe(3,5-di-F), L-Phe(3-CF3), L-Phe(3-OMe), L-Phe(4-Cl), L-Phe(4- CONH2), L-homohomoPhe, L-homoSer(2-Me,4-Cl-phenyl), L-homoTyr, L-Trp, e), L-Trp(5-OH), L-Trp (6-OMe), L-Trp(7-F), L-Trp(7-Me),
-Trp(Me), L-Tyr, L-Tyr(Bn), and L-Tyr(Me);
R10 group is selected from sidechain of the following amino acids: L-Asp, L- Phe(4-COOH). and L-Tyr(CH2COOH);
R11 group is selected from sidechain of the following amino acids: L-l-Nal. L-2- Nal, L-Ala(P-indol-4-yl), L-Phe(3-OMe), L-Phe(4-NH2), L-Trp, L-Trp(5-OH). L- Trp(Me), and L-Tyr;
R12 group is selected from sidechain of the following amino acids: Aib,
L-Asp, D-Ala, D-Asp, D-Glu, D-Phe, D-His, D-Lys, D-Leu, D-Asn, D-Gln, D-Arg, D- Ser, D-Thr, D-Trp, D-Tyr, L-Glu, Gly, L-Ser, L-Trp(l-CH2COOH), and L- Tyr(CH2COOH):
R17a is each independently selected from hydrogen, CH?. -CH2OH, -(CH2)I-4NH2. -(CH2)1-4NH-C(=NH)NH2. -CH2CO2H, -(CH2)2CO2H,
R17 is each independently selected from hydrogen. -CH?, -(CH2)ZNH2, - (C(R17a)2)o-4-X’-R31, -(CH2)ZCO2H, -CH2OH, CH2C CH, and -(CH2)i-3-triazolyl-X’-R35, and R35 is selected from -CO2H, -C(O)NH2, CH3, tetrazole, and lithocolic acid and analogs; provided that at least one R17 is other than hydrogen, -CH?, or -CH2OH;
In another embodiment, compounds of formula (I), wherein,
R1 group is selected from sidechain of the following amino acids: L-Phe(4-COPh) (L-Bpa), L-Phe, L-Phe(3,4,5-tri-F), L- L-Phe(3,5-di-F), L-Phe(3-CN), L-Phe(4-CN), L- Phe(4-CONH2), L-Phe(4-COOH), L-Phe(3-CONH2), L-Tyr(benzyl), and L- Tyr(CH2COOH):
R2 group is L-Val;
R3 group is selected from sidechain of the following amino acids: L-alloT, L-Asp, L-Trp, L-Trp(l-CH2COOH), L-Trp(5-OBn), and L-Tyr;
R’ group is selected from sidechain of the following amino acids: L-Asp, L-Dab, L-Dab(COCH2NH2), D-His, D-Arg, Gly, L-Hyp, and L-Tyr(CH2COOH);
R6 group is selected from sidechain of the following amino acids: L-2-Nal, L- Bip(3'-COOH), L-Bip(4'-COOH), L-Phe(4-Cl), L-Phe(4-CH2NH2), L-Phe(4-CONH2), L-
Phe(4-COOH), L-Trp, L-Trp(l-CH2COOH), L-Trp(5-O-benzyl), L-Tyr,
L-Tyr(CH2C00H), L-Tyr(phenyl), and L-Tyr(propargy 1);
R7 group is selected from sidechain of the following amino acids: L-Dap, L- Phe(4-COOH). L-Asn. L-Ser, L-Thr, L-Tyr. and L-Tyr(propargyl);
R8 group is selected from sidechain of the following amino acids: L-Bip, L-Bip(2'- COOMe), L-Bip(2'-Et,4'-OH), L-Bip(2'-F), L-Bip(2'-Me), L-Bip(2-O-nBu), L-Bip(2'- OMe,4'-F), L-Bip(3',5'-di-F), L-Bip(3-CN), L-Bip(3'-COOH), L-Bip(3'-Me), L-Bip(4'- Cl). L-Bip(4'-CONMe2), L-Bip(4'-COOH), L-Bip(4'-NHCOMe), L-Bip(4'-OPh), L- Phe(4-(2-benzofuranyl)). L-Phe(4-(2-(3-CN -thienyl)). L-Phe(4-CH=CH-Ph), and L- Tyr(Ph);
R9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Ala(P-quinolin-6-yl), L-Ala(P-isoquinolin-8-yl), L-Ala(P-isoquinolin-4-yl), L-Ala(P- quinolin-3-yl), L-Ala(P-3-methoxy-isoquinolin-7-yl), L-Bzt, L-Phe, L-Phe(2-Cl), L- Phe(3,4-di-F), L-Phe(3,5-di-F), L-Phe(3-CF2), L-Phe(3-OMe), L-Phe(4-Cl), L-Phe(4- CONH2), L-homohomoPhe, L-homoSer(2-Me,4-Cl-phenyl), L-homoTyr, L-Trp,
R10 group is L-Asp;
R11 group is selected from sidechain of the following amino acids: L-Ala(P-indol- 4-yl), L-Trp, L-Trp(5-OH), and L-Tyr;
R12 group is selected from sidechain of the following amino acids:
D-Trp, Gly, and L-Ser,;
In another embodiment, compounds of formula (I), (<100 nM), wherein,
R1 group is selected from sidechain of the following amino acids: L-Phe, L- Phe(3,5-di-F), L-Phe(4-CONH2), L-Phe(4-COOH), and L-Phe(3-CONH2);
R3 group is selected from sidechain of the following amino acids: L-Asp. L-Trp, L-Trp(l-CH2COOH), and L-Tyr;
R’ group is selected from sidechain of the following amino acids: L-Asp, D-Arg, and Gly;
R6 group is selected from sidechain of the following amino acids: L-Phe(4- COOH), L-Trp, and L-Tyr;
R7 group is L-Tyr;
R8 group is selected from sidechain of the following amino acids: L-Bip, L-Bip(2'- Me), L-Bip(3'-COOH), L-Bip(4'-CONMe2), L-Bip(4'-COOH), and L-Bip(4'-NHCOMe);
R9 group is selected from sidechain of the following amino acids: L-l-Nal, L- Phe(3-CFs), L-Trp, and L-Tyr;
R10 group is L-Asp;
R11 group is selected from sidechain of the following amino acids: L-Trp and L- Tyr;
R12 group is selected from sidechain of the following ammo acids: D-Ser. and Gly;
As shown below in Tables 1-7. the following compounds of the invention show activity at less than or equal to 1.0 nM in the assay included later in the application.
Table 1
Table 3
Table 4
Table 5
Table 6
Table 7
Definitions
The definitions provided herein apply, without limitation, to the terms as used throughout this specification, unless otherwise limited in specific instances.
Those of ordinary skill in the art of amino acid and peptide chemistry are aware that an amino acid includes a compound represented by the general structure:
where R and R' are as discussed herein.
Unless otherwise indicated, the term "amino acid" as employed herein, alone or as part of another group, includes, without limitation, an amino group and a carboxyl group linked to the same carbon, referred to as "a" carbon, where R and/or R' can be a natural or an un-natural side chain, including hydrogen. The absolute "S" configuration at the "a" carbon is commonly referred to as the "L" or "natural" configuration. In the case where both the "R" and the "R'"(prime) substituents equal hydrogen, the amino acid is glycine and is not chiral.
The term “naturally occurring amino acid side chain." as used herein, refers to side chain of any of the naturally occurring amino acids (i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, -histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, t rosine, and valine) usually in the S -configuration (i.e., the L-amino acid).
The term “non-naturally occurring amino acid side chain,” as used herein, refers to a side chain of any naturally occurring amino acid usually in the R-configuration (i.e.. the D-amino acid) or to a group other than a naturally occurring amino acid side chain in R- or S-configuration (i.e., the D- or L-amino acid, respectively) selected from:
The "inhibitory concentration" of LAG-3 inhibitor is intended to mean the concentration at which a compound screened in an assay of the disclosure inhibits a measurable percentage of the interaction of LAG-3 with MHC Class II molecules. Examples of "inhibitory concentration" values range from IC50 to IC90, and are preferably, IC50, ICeo, IC70, ICso, or IC90, which represent 50%, 60%, 70%, 80% or 90% reduction in LAG-3/MHC Class II molecules binding activity, respectively. More preferably, the "inhibitory concentration" is measured as the IC50 value. It is understood that another designation for IC50 is the half-maximal inhibitor}' concentration.
Binding of the macrocyclic peptides to LAG-3 can be measured, for example, by methods such as homogeneous time-resolved fluorescence (HTRF), Surface Plasmon Resonance (SPR), isothermal titration calorimetry (ITC), nuclear magnetic resonance spectroscopy (NMR), and the like. Further, binding of the macrocyclic peptides to LAG- 3 expressed on the surface of cells can be measured as described herein in cellular binding assays.
Administration of a therapeutic agent described herein includes, without limitation, administration of a therapeutically effective amount of therapeutic agent. The term "therapeutically effective amount" as used herein refers, without limitation, to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the LAG-3/MHC Class II molecules binding inhibitors described herein. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example and without limitation, treatment or prevention of the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance.
In another aspect, the disclosure pertains to methods of inhibiting growth of tumor cells in a subject using the macrocyclic peptides of the present disclosure. As demonstrated herein, the macrocyclic peptides of the present disclosure are capable of binding to LAG-3, disrupting the interaction between LAG-3 and MHC class II molecules. As a result, the macrocyclic peptides of the present disclosure are potentially
useful for modifying an immune response, treating diseases such as cancer or infectious disease, stimulating a protective autoimmune response or to stimulate antigen-specific immune responses (e.g., by coadministration of LAG-3 blocking peptides with an antigen of interest).
In order that the present disclosure may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description.
The terms "Programmed Death Ligand 1", "Programmed Cell Death Ligand 1", "Protein PD-L1". "PD-L1", "PDL1". "PDCDL1", "hPD-Ll", "hPD-LI", "CD274" and "B7-H1" are used interchangeably, and include variants, isoforms, species homologs of human PD-L1, and analogs having at least one common epitope with PD-L1. The complete PD-L1 sequence can be found under GENBANK® Accession No. NP_054862.
The terms "Programmed Death 1", "Programmed Cell Death 1", "Protein PD-1", "PD-1", "PD1", "PDCD1". "hPD-1" and "hPD-I" are used interchangeably, and include variants, isoforms, species homologs of human PD-1, and analogs having at least one common epitope with PD-1. The complete PD-1 sequence can be found under GENBANK® Accession No. U64863.
The terms "cytotoxic T lymphocyte-associated antigen-4", "CTLA-4", "CTLA4". "CTLA-4 antigen" and "CD152" (see, e.g., Murata, Am. J. Pathol., 155:453-460 (1999)) are used interchangeably, and include variants, isoforms, species homologs of human CTLA-4, and analogs having at least one common epitope with CTLA-4 (see, e.g., Balzano, Int. J. Cancer Suppl., 7:28-32 (1992)). The complete CTLA-4 nucleic acid sequence can be found under GENBANK® Accession No. L15006.
The term "immune response" refers to the action of. for example, lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including macrocyclic peptides, cytokines, and complement) that results in selective damage to, destruction of, or elimination from the human body of invading pathogens, cells or tissues infected with pathogens, cancerous cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
A "signal transduction pathway" refers to the biochemical relationship between a variety of signal transduction molecules that play a role in the transmission of a signal from one portion of a cell to another portion of a cell. As used herein, the phrase "cell surface receptor" includes, for example, molecules and complexes of molecules capable of receiving a signal and the transmission of such a signal across the plasma membrane of a cell. An example of a "cell surface receptor" of the present disclosure is the PD-1 receptor.
The term "macrocyclic peptide derivatives" refers to any modified form of the macrocyclic peptides disclosed herein, e.g., mutations, isofonns, peptides with altered linker backbones, conjugates with an antibody and/or another agent, etc..
As used herein, a (preferred?) macrocyclic peptide of the present disclosure that "specifically binds to human LAG-3" is intended to refer to a macrocyclic peptide that binds to human LAG-3 with an IC50 of less than about 1000 nM, less than about 300 nM, less than about less than about 100 nM, less than about 80 nM, less than about 60 nM, less than about 40 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 1 nM. or less. In this context, the term "about" shall be construed to mean anywhere between ± 1, 2, 3, 4. 5, 6, 7. 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nM more or less than the cited amount.
The term "treatment" or "therapy" refers to administering an active agent with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect a condition (e.g. , a disease), the symptoms of the condition, or to prevent or delay the onset of the symptoms, complications, biochemical indicia of a disease, or otherwise arrest or inhibit further development of the disease, condition, or disorder in a statistically significant manner.
As used herein, "about" or "comprising essentially of mean within an acceptable error range for the particular value as determined by one of ordinary’ skill in the art, which will depend in part on how the value is measured or determined, z.e., the limitations of the measurement system. For example, "about" or "comprising essentially of can mean within one or more than one standard deviation per the practice in the art. Alternatively, "about" or "comprising essentially of can mean a range of up to 20%. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an
order of magnitude or up to 5-fold of a value. When particular values are provided in the application and claims, unless otherwise stated, the meaning of "about" or "comprising essentially of should be assumed to be within an acceptable error range for that particular value.
As described herein, any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
Variant Macrocyclic Peptides
In yet another embodiment, a macrocyclic peptide of the disclosure comprises amino acid sequences that are homologous to the amino acid sequences of the macrocyclic peptides described herein, and wherein the macrocyclic peptides retain the desired functional and/or biological properties of the macrocyclic peptide of the disclosure.
For example, the disclosure provides a macrocyclic peptide, or antigen-binding portion thereof, comprising: an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the compounds described herein; and the macrocyclic peptide exhibits one or more of the following properties:
(a) the macrocyclic peptide binds to human LAG-3 with an IC50 of 200 nM or less;
(b) the macrocyclic peptide does not substantially bind to human CD4;
(c) the macrocyclic peptide binds to human LAG-3 and one or more of the following: cynomolgus monkey LAG-3, and/or mouse LAG-3;
(d) the macrocyclic peptide inhibits the binding of LAG-3 to MHC Class II moleucules;
(e) the macrocyclic peptide inhibits tumor cell growth in a cellular assay and/or in vivo assay;
In other embodiments, the macrocyclic peptide amino acid sequences may be about 80%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about
91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or about 99% homologous to the sequences set forth above. In this context, the term "about" shall be construed to mean anywhere between 1, 2, 3, 4, or 5 percent more or less than the cited amount. A macrocyclic peptide of the present disclosure having sequences with high identity (z.e., 80% or greater) to the sequences set forth above, can be obtained by mutating the sequences during chemical synthesis, for example, followed by testing of the altered macrocyclic peptide for retained function (i.e.. the functions set forth in (a) through (i) above) using the functional assays described herein. The biological and/or functional activity of the variant macrocyclic peptide amino acid sequences may be at least about lx, 2x, 3x, 4x, 5x, 6x,7x, 8x, 9x, or lOx more than the reference macrocyclic peptide on which the variant is based. In this context, the term "about" shall be construed to mean anywhere between O. lx, 0.2x, 0.3x, 0.4x, 0.5x, 0.6x, 0.7x, 0.8x, or 0.9x more or less than the cited amount.
As used herein, the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % homology = # of identical positions / total # of positions.times. 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
The percent identity between two amino acid sequences can be determined using the algorithm of Meyers E. et al., (Comput. Appl. Biosci., 4: 11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman et al. (J. Mol. Biol., 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG® software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8. 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
Macrocyclic Peptides with Conservative Modifications
In yet another embodiment, a macrocyclic peptide of the disclosure comprises amino acid sequences that are homologous to the amino acid sequences of the macrocyclic peptides described herein, and wherein the macrocyclic peptides retain the desired functional and/or biological properties of the macrocyclic peptide of the disclosure.
For example, the disclosure provides a macrocyclic peptide, or antigen-binding portion thereof, comprising: an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the macrocyclic peptides described herein, wherein one or more amino acids have been substituted with a conservative amino acid; and the macrocyclic peptide exhibits one or more of the following properties:
(a) the macrocyclic peptide binds to human LAG-3 with an IC50 of 200 nM or less;
(b) the macrocyclic peptide does not substantially bind to human CD4;
(c) the macrocyclic peptide binds to human LAG-3 and one or more of the following: cynomolgus monkey LAG-3, and/or mouse LAG-3;
(d) the macrocyclic peptide inhibits the binding of LAG-3 to MHC Class II moleucules;
(e) the macrocyclic peptide inhibits tumor cell growth in a cellular assay and/or in vivo assay;
As used herein, the term "conservative sequence modifications" is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the macrocyclic peptide containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the disclosure by standard techniques know n in the art, such as substitution of peptide amidites during chemical synthesis, site- directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side
chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g, glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g. tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the antigen binding regions of macrocyclic peptides of the disclosure can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i. e. , the functions set forth in (a) thru (i) above) using the functional assays described herein. Conservative amino acid substitutions may also be selected from one or more non-naturally occurring amino acids disclosed herein.
Peptide Synthesis
The description of the present disclosure herein should be construed in congruity with the laws and principals of chemical bonding. It should be understood that the compounds encompassed by the present disclosure are those that are suitably stable for use as pharmaceutical agent. One of skill in the art will know what compounds would and would not be stable based on the general principles of chemical bonding and stability'.
The macrocyclic peptides of the present disclosure can be produced by methods known in the art, such as they can be synthesized chemically, recombinantly in a cell free system, recombinantly within a cell or can be isolated from a biological source. Chemical synthesis of a macrocyclic peptide of the present disclosure can be carried out using a variety of art recognized methods, including stepwise solid phase synthesis, semisynthesis through the conformationally-assisted re-ligation of peptide fragments, enzymatic ligation of cloned or synthetic peptide segments, and chemical ligation. A preferred method to synthesize the macrocyclic peptides and analogs thereof described herein is chemical synthesis using various solid-phase techniques such as those described in Chan, W.C. et al, eds., Fmoc Solid Phase Synthesis, Oxford University Press, Oxford (2000); Barany, G. et al, The Peptides: Analysis. Synthesis, Biology, Vol. 2 : "Special Methods in Peptide Synthesis, Part A", pp. 3-284, Gross, E. et al, eds., Academic Press, New York (1980); in Atherton, E., Sheppard, R. C. Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, Oxford, England (1989); and in Stewart, J. M. Young, J.
D. Solid-Phase Peptide Synthesis, 2nd Edition, Pierce Chemical Co., Rockford, IL (1984). The preferred strategy is based on the (9-fluorenylmethyloxy carbonyl) group (Fmoc) for temporary protection of the a-amino group, in combination with the /e/7-but l group (®u) for temporary protection of the amino acid side chains (see for example Atherton. E. et al, "The Fluorenylmethoxy carbony l Amino Protecting Group", in The Peptides: Analysis. Synthesis, Biology. Vol. 9 : "Special Methods in Peptide Synthesis. Part C", pp. 1-38, Undenfriend, S. et al, eds.. Academic Press, San Diego (1987).
The peptides can be synthesized in a stepwise manner on an insoluble polymer support (also referred to as "resin") starting from the C-terminus of the peptide. A synthesis is begun by appending the C-terminal amino acid of the peptide to the resin through formation of an amide or ester linkage. This allows the eventual release of the resulting peptide as a C-terminal amide or carboxylic acid, respectively.
The C-terminal amino acid and all other amino acids used in the synthesis are required to have their a-amino groups and side chain functionalities (if present) differentially protected such that the a-amino protecting group may be selectively removed during the synthesis. The coupling of an amino acid is performed by activation of its carboxyl group as an active ester and reaction thereof with the unblocked a-amino group of the N-terminal amino acid appended to the resin. The sequence of a-amino group deprotection and coupling is repeated until the entire peptide sequence is assembled. The peptide is then released from the resin with concomitant deprotection of the side chain functionalities, usually in the presence of appropriate scavengers to limit side reactions. The resulting peptide is finally purified by reverse phase HPLC.
The synthesis of the peptidyl-resins required as precursors to the final peptides utilizes commercially available cross-linked polystyrene polymer resins (Novabiochem, San Diego, CA; Applied Biosystems, Foster City, CA). Preferred solid supports are: 4- (2'.4'-dimethoxyphenyl-Fmoc-aminomethyl)-phenoxyacetyl-p-methyl benzhydrylamine resin (Rink amide MBHA resin); 9-Fmoc-amino-xanthen-3-yloxy-Merrifield resin (Sieber amide resin); 4-(9-Fmoc)aminomethyl-3,5- dimethoxyphenoxy)valerylaminomethyl-Merrifield resin (PAL resin), for C-terminal carboxamides. Coupling of first and subsequent amino acids can be accomplished using HOBt, 6-Cl-HOBt or HO At active esters produced from DIC/HOBt, HBTU/HOBt, BOP, PyBOP, or from DIC/6-Cl-HOBt, HCTU, DIC/HOAt or HATU, respectively. Preferred
solid supports are: 2-chlorotrityl chloride resin and 9-Fmoc-amino-xanthen-3-yloxy- Merrifield resin (Sieber amide resin) for protected peptide fragments. Loading of the first amino acid onto the 2-chlorotrityl chloride resin is best achieved by reacting the Fmoc- protected amino acid with the resin in dichloromethane and DIEA. If necessary', a small amount of DMF may be added to solubilize the amino acid.
The syntheses of the peptide analogs described herein can be carried out by using a single or multi-channel peptide synthesizer, such as an CEM Liberty Microwave synthesizer, or a Protein Technologies, Inc. Prelude (6 channels) or Symphony (12 channels) or Symphony X (24 channels) synthesizer.
The peptidyl-resin precursors for their respective peptides may be cleaved and deprotected using any standard procedure (see, for example, King, D.S. et aL Int. J. Peptide Protein Res., 36:255-266 (1990)). A desired method is the use of TFA in the presence of water, TIS as scavenger, and DTT or TCEP as the disulfide reducing agent. Typically, the peptidyl-resin is stirred in TFA/water/TIS/DTT (94:3:3: 1), v:v:v:w; 1 mL/100 mg of peptidyl resin) for 1.5-3 hrs at room temperature. The spent resin is then filtered off and the TFA solution was cooled and Et20 solution was added. The precipitates were collected by centrifuging and decanting the ether layer (3 x). The resulting crude peptide is either redissolved directly into DMF or DMSO or CH3CN/H2O for purification by preparative HPLC or used directly in the next step.
Peptides with the desired purity can be obtained by purification using preparative HPLC, for example, on a Waters Model 4000 or a Shimadzu Model LC-8A liquid chromatography. The solution of crude peptide is injected into a YMC S5 ODS (20 x 100 mm) column and eluted with a linear gradient of MeCN in water, both buffered with 0.1% TFA, using a flow rate of 14-20 mL/min with effluent monitoring by UV absorbance at 220 nm. The structures of the purified peptides can be confirmed by electro-spray MS analysis.
Useful Fmoc amino acids derivatives are shown below.
Examples of Orthogonally Protected Amino Acids used in Solid Phase Synthesis
Analytical Data:
Mass Spectrometry: “ESI-MS(+)” signifies electrospray ionization mass spectrometry performed in positive ion mode; “ESI-MS(-)” signifies electrospray ionization mass spectrometry’ performed in negative ion mode; L‘ESI-HRMS(+)” signifies high-resolution electrospray ionization mass spectrometry performed in positive ion mode; ‘'ESI-HRMS(-)” signifies high-resolution electrospray ionization mass spectrometry’ performed in negative ion mode. The detected masses are reported following the “m/z' unit designation. Compounds with exact masses greater than 1000 were often detected as double-charged or triple-charged ions.
The crude material was purified via preparative LC/MS. Fractions containing the desired product were combined and dried via centrifugal evaporation.
Analytical LC/MS Condition A:
Column: Waters Acquity UPLC BEH C18, 2. 1 x 50 mm, 1.7-pm particles; Mobile Phase A: 5:95 acetonitrile: water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile: water with 10 mM ammonium acetate; Temperature: 50 °C; Gradient: 0- 100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
Analytical LC/MS Condition B:
Column: Waters Acquity UPLC BEH C18, 2. 1 x 50 mm, 1.7-|im particles; Mobile Phase A: 5:95 acetonitrile: water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile: water with 0.1% trifluoroacetic acid; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
Analytical LC/MS Condition C: Column: Waters Acquity UPLC BEH Cl 8, 2.1 x 50 mm, 1.7-pm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate;
Temperature: 70 °C; Gradient: 0-100% B over 3 minutes, then a 2.0-minute hold at 100% B; Flow: 0.75 mL/min; Detection: UV at 220 nm.
Analytical LC/MS Condition D: Column: Waters Acquity UPLC BEH C 18, 2.1 x 50 mm, 1.7-pm particles; Mobile Phase A: 5:95 acetonitrile:water with 0. 1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 70 °C; Gradient: 0-100% B over 3 minutes, then a 2.0-minute hold at 100% B; Flow: 0.75 mL/min; Detection: UV at 220 nm.
Analytical LC/MS Condition E: Column: Kinetex XB C18, 3.0 x 75 mm, 2.6-pm particles; Mobile Phase A: 10 mM ammonium formate in water: acetonitrile (98:2); Mobile Phase B: 10 mM ammonium formate in Water: acetonitrile (02:98); Gradient: 20- 100% B over 4 minutes, then a 0.6-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 254 nm.
Analytical LC/MS Condition F: Column: Ascentis Express C18, 2.1 x 50 mm, 2.7-pm particles; Mobile Phase A: 10 mM ammonium acetate in water: acetonitrile (95:5); Mobile Phase B: 10 mM ammonium acetate in Water: acetonitrile (05:95), Temperature: 50 °C; Gradient: 0-100% B over 3 minutes; Flow: 1.0 mL/min; Detection: UV at 220 nm.
Analytical LC/MS Condition G: Column: X Bridge C18, 4.6 x 50 mm, 5-qm particles; Mobile Phase A: 0.1% TFA in water; Mobile Phase B: acetonitrile, Temperature: 35 °C; Gradient: 5-95% B over 4 minutes; Flow: 4.0 mL/min; Detection: UV at 220 nm.
[0001] The following abbreviations are employed in the Examples and elsewhere herein:
Ph = phenyl
mmol = millimole(s) meq = milliequivalent rt or RT = room temperature sat or sat'd = saturated aq. = aqueous mp = melting point
BOP reagent = benzotriazol- 1-yloxy-tris-dimethylamino-phosphonium hexafluorophosphate (Castro's reagent)
PyBOP reagent = benzotriazol-l-yloxy-tripyrrolidino phosphonium hexafluorophosphate HBTU = 2-( I H-Benzotriazol- 1 -yl)- 1. 1.3.3-tetramethyluronim hexafluorophosphate HATU = 0-(7-Azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronim hexafluorophosphate
HCTU = 2-(6-Chloro-l-H-benzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
T3P = 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide
DMAP = 4-(dimethylamino)pyridine
DIEA = diisopropylethylamine
Fmoc or FMOC = fluorenylmethyloxy carbonyl
Boc or BOC = tert-butyloxycarbonyl
HOBT or HOBT*H2O = 1 -hydroxy benzotriazole hydrate
Cl-HOBt = 6-Chloro-benzotriazole
HO AT = l-hydroxy-7-azabenzotriazole
HPLC = high performance liquid chromatography
LC/MS = high performance liquid chromatography /mass spectrometry
MS or Mass Spec = mass spectrometry
NMR = nuclear magnetic resonance
Sc or SC or SQ = sub-cutaneous
IP or ip = intra-peritoneal
General Procedures:
All manipulations were performed under automation on a Prelude Prelude, or a Symphony, or Symphony X peptide synthesizer (Protein Technologies). All procedures were performed according to the published methods (e.g., WO 2023/225661).
Prelude: Resin-swelling procedure, Single-coupling procedure, Single-coupling extended time procedure, Chloroacetic Anhydride coupling. Single-Coupling Manual Addition Procedure A, Single-Coupling Manual Addition Procedure B, Manual removal ofFmoc group procedure:
Symphony: Resin-swelling procedure, Single-coupling procedure, Single-coupling extended time procedure. Double-coupling extended time procedure, Chloroacetic Anhydride coupling:
Symphony X: Resin-swelling procedure, Single-coupling procedure, Single-coupling 3 deprotections procedure, Single-coupling extended time procedure, Single-coupling 3 deprotections extended time procedure, Pre-activated single-coupling procedure. SingleCoupling Manual Addition Procedure A, Single-Coupling Manual Addition Procedure B:
Chloroacetic Anhydride coupling, Final rinse and dry procedure.
The following procedures were performed according to the published methods (e.g., WO 2023/225661).
Global Deprotection Method. Cyclization Method, N-Methylation on-Resin Method A, N- Methylation On-resin Method B (Turner, R.A. et al, Org. Lett., 15(19):5012-5015 (2013)), N-Alkylalion On-resin Procedure Method A, N-Alkylalion On-resin Procedure Method B, N-Nosylate Formation Procedure, N-Nosylate Removal Procedure, General Procedure for Preloading amines on the PL-FMP resin, General Procedure for Preloading Fmoc-Amino Acids on Cl-trityl resin, Click Reaction On-Resin Method A, Click Reaction On-Resin Method B, Suzuki Reaction On-resin Procedure, Fatty: acid chain coupling procedure A, Fatty acid chain coupling procedure B, General Purification Procedures.
Unnatural Fmoc-Amino Acid Synthesis and fatty acid tails were prepared according to the published methods (e.g., WO 2023/225661).
Preparation of INT-1000
INT-1000 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ), and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
“Symphony Method B: Resin-swelling procedure” was followed;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Lys(Boc)- OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly- OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly- OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Cys(Trt)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc- Asp(OtBu)-OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Bip-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-
OH;
“Symphony Method B: Double-coupling procedure’" was followed with Fmoc-Asp(Boc)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc- Asp(OtBu)-OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Val-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Phe-OH followed by “Manual removal of Fmoc group procedure”; “Chloroacetic acid coupling procedure B” was followed; “Deprotection Method B” was followed; “Cyclization Method B” was followed.
The crude material was purified via preparative LC/MS with the following conditions: Column: Waters XBridge C18, 19 x 150 mm, 5 pm particles; Mobile Phase A: 5:95 acetonitrile: water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile: water with 0.1% trifluoroacetic acid; Gradient: 10-60% B over 20 minutes, then a 2-minute hold at 100% B; Flow: 40 mL/min.
The yield of the product was 4.8 mg, and its estimated urity by LCMS analysis was 83.6%.
Analysis condition B: Retention time = 1.39 min; ESI-MS(+) m/z [M+2H]2' : 1097.9.
Preparation ofINT-1001
INT-1001 was prepared, using Sieber resin on a 0.025 mmol scale, following the general synthetic sequence described for the preparation of Example 1000. The yield of the product was 2.6 mg, and its estimated purity by LCMS analysis was 97. 1%. Analysis condition A: Retention time = 1.53 min; ESI-MS(+) m/z [M+2H]2+: 1080.2.
INT-1002 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ) and the following procedures were done sequentially: “Tail loading procedure B” was followed with Fmoc-Lys(Boc)-OH, “Manual removal of Fmoc group procedure C” was followed, “Linker coupling procedure” was followed with Fmoc-12-amino-4,7,10-trioxadodecanoic acid, and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
“Symphony Method B: Resin-swelling procedure” was followed;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Cys(Trt)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc- Asp(OtBu)-OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method B: Double-coupling procedure"’ was followed with Fmoc-Bip-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Gly-OH; “Symphony Method B: Double-coupling procedure” was followed with Fmoc- Asp(OtBu)-OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method B: Double-coupling procedure” was followed with Fmoc-Val-OH; "‘Symphony Method B: Double-coupling procedure” was followed with Fmoc-Phe-OH; “Chloroacetic acid coupling procedure B” was followed; “Deprotection Method B” was followed; “Cyclization Method B” was followed. The crude material was purified via preparative LC/MS The yield of the product was 2.8 mg, and its estimated purity by LCMS analysis was 91%. Analysis condition A: Retention time = 1.56 min; ESI-MS(+) m/z [M+2H]2+: 1096.0.
Preparation of INT-1003
INT-1003 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol and the following procedures were done sequentially: “Tail loading procedure B” was followed with Fmoc-Lys(Boc)-OH, “Manual removal of Fmoc group procedure C” was followed, “Linker coupling
procedure’' was followed with Fmoc-12-amino-4,7,10-trioxadodecanoic acid, and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
“Symphony Method A: Resin-swelling procedure” was followed;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Cys(Trt)- OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc-Gly-OH; “Symphony Method A: Single -coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc- Asp(OtBu)-OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc-Bip-OH; “Symphony Method A: Single -coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single -coupling procedure” was followed with Fmoc-Gly-OH;
“Symphony Method A: Single -coupling procedure” was follow ed with Fmoc- Asp(OtBu)-OH;
“Symphony Method A: Single -coupling procedure” w as follow ed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single -coupling procedure” was follow ed with Fmoc-Val-OH; “Symphony Method A: Single-coupling followed by removal of Fmoc group procedure” was followed with (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(4-(tert- butoxycarbonyl)phenyl)propanoic acid; “Chloroacetic acid coupling procedure A” was followed:
“Deprotection Method A” was followed; “Cyclization Method A” was followed.
The crude material was purified via preparative LC/MS. The yield of the product was 1.7 mg, and its estimated purity by LCMS analysis was 90%.
Analysis condition A: Retention time = 1.01 min; ESI-MS(+) m'z [M+2H]2+; 1095.0.
Analysis condition B: Retention time = 1.28 min; ESI-MS(+) m'z [M+2H]2+: 1095.1.
INT-1004 to INT-1060 were prepared following similar procedures described for INT- 1000 to INT-1003 and general synthetic and purification procedures
Preparation of Example 1001
Example 1001 was prepared as follows; to a 25-mL polypropylene solid-phase reaction vessel was added Sieber resin (70 mg, 0.05 mmol ), and the reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially:
“Symphony Method A: Resin-swelling procedure'’ was followed;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Gly-OH; “Symphony Method A: Single-coupling procedure” was followed with Fmoc-Cys(Trt)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Gly-OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Asp(OtBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Trp(Boc)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Bip-OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Lys(Boc)- OH;
“Symphony Method A: Single-coupling procedure’' was followed with Fmoc-Asp(OtBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Tyr(tBu)- OH;
“Symphony Method A: Single-coupling procedure” was followed with Fmoc-Val-OH; “Symphony Method A: Single-coupling procedure” was followed with Fmoc-Phe-OH followed by “Manual removal of Fmoc group procedure”; “Chloroacetic acid coupling procedure A” was followed; “Deprotection Method A” was followed; “Cyclization Method A” was followed.
“Cyclization Method A” was followed.
“Fatty acid chain coupling procedure C” was followed with perfluorophenyl palmitate; The crude material was purified via preparative LC/MS with the following conditions: Column: Waters X-bridge C18, 30 x 150 mm, 5-um particles: Mobile Phase A: 5:95 acetonitrile: water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile: water with 0.1% TFA. Gradient: 10-60% B over 20 minutes, then a 5 minute hold at 100% B: Flow: 45 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation.
The yield of the product was 3.1 mg, and its estimated purity by LCMS analysis was 97%.
Analysis condition A: Retention time = 2.19 min. Analysis condition B: Retention time = 2.49 min; ESI-MS(+) m/z [M+2H]2+: 1090.1.
Preparation of Example 1002
Example 1002 was prepared as such: in a 25 mL glass solid support chemistry reaction vessel equipped with a frit were performed the following procedures sequentially:
“Tail loading procedure A” was followed with Fmoc-L-Glu(2-phenylisopropyloxy)-OH; “Manual removal of Fmoc group procedure C” was followed;
“Fatty acid chain coupling procedure A” was followed with pamitic acid;
“Linker coupling procedure’’ w as followed with l-(9-fluorenylmethyloxycarbonyl- amino)-4,7,10-trioxa-13-tridecanamine hydrochloride;
“Manual removal of Fmoc group procedure C” was followed;
“Manual coupling procedure D” was followed with Fmoc-Cys(Trt)-OH;
The resin w as then transferred to a 45 mL polypropylene solid-phase reaction vessel was added the previously modificated Wang resin (646 mg, 0.250 mmol ), and the reaction vessel was placed on the Prelude peptide synthesizer. The following procedures were then performed sequentially:
“Prelude Method C: Resin-swelling procedure” was followed;
“Prelude Method C: Double-coupling procedure” w as followed with Fmoc-Gly-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Asp(OtBu)- OH;
“Prelude Method C: Double-coupling procedure” w as followed with Fmoc-Trp(Boc)-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Bip-OH;
“Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH;
“Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-D-Arg(Pbf)- OH;
“Prelude Method C: Double-coupling procedure” w as followed with Fmoc-Asp(OtBu)- OH;
“Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH; “Prelude Method C: Double-coupling procedure” was followed with Fmoc-Val-OH;
“Prelude Method C: Double-coupling procedure” was followed with Fmoc-Tyr(tBu)-OH; “Manual removal of Fmoc group procedure C” was followed;
“Manual coupling of Fmoc group procedure C” was followed with (S)-2-((((9H-fluoren- 9-yl)methoxy)carbonyl)amino)-3-(4-(tert-butoxycarbonyl)phenyl)propanoic acid;
“Chloroacetyl Chloride coupling procedure A” was followed;
“Deprotection Method C” was followed; “Cyclization Method C” was followed. The crude material was purified via preparative LC/MS. The yield of the product was 31.3 mg, and its estimated purity by LCMS analysis was 98%. Analysis condition A: Retention time = 1.78 min: ESI-MS(+) m/z [M+3HJ3+: 843.3. Analysis condition B: Retention time = 2.22 min; ESI-MS(+) m/z [M+3H]3? 843.5.
The following examples were prepared following the general synthetic sequence described for the preparation of Examples 1001 and 1002,
Preparation of Example 1003
Preparation of Example 1004
Preparation of Example 1005
Preparation of Example 1006
Preparation of Example 1007
Preparation of Example 1008
Preparation of Example 1009
Preparation of Example 1010
Preparation of Example 1011
Preparation of Example 1012
Preparation of Example 1013
Preparation of Example 1014
Preparation of Example 1015
Preparation of Example 1016
Preparation of Example 1017,
Preparation of Example 1018
Preparation of Example 1019
Preparation of Example 1020
Preparation of Example 1021
Preparation of Example 1022
Preparation of Example 1023
Preparation of Example 1024
Preparation of Example 1025
Preparation of Example J 026
Preparation of Example 1027
Preparation of Example 1028
Preparation of Example 1029
Example 1029 was prepared as follows; to a 25 mL polypropylene solid-phase reaction vessel was added 2-Chlorotrityl resin pre-loaded with (S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)pent-4-ynoic acid and the following procedures were done sequentially: '‘Tail loading procedure C” was followed with (S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)pent-4-ynoic acid;
The reaction vessel was placed on the Symphony peptide synthesizer. The following procedures were then performed sequentially: “Symphony Method B: Resin-swelling procedure” was followed; “Symphony Method B: Double-coupling procedure” was followed
"Chloroacetic acid coupling procedure B" was followed; "Deprotection Method B" was followed: “Click tail cycloaddition procedure” was followed with (S)-tert-butyl 1-azido- 22-(tert-butoxy carbonyl)- 19.24-dioxo-3.6.9.12, 15 -pentaoxa- 18.23-diazahentetracontan- 41-oate and 25% water in tert-butanol; The crude material was purified via preparative LC/MS. The yield of the product was 1.6 mg, and its estimated purity7 by LCMS analysis was 98%.
Analysis condition A: Retention time = 1.60 min; ESI-MS(+) m/z [M+2H]2+: 1394.4. Analysis condition B: Retention time = 2.03 min; ESI-MS(+) m/z [M+2H]2+: 1394.4.
The following Examples were prepared, using 2-Chlorotrityl resin pre-loaded with Fmoc-NH-fatty acid following the general synthetic sequence described for the preparation of INT-1001.
Preparation of Example 1031
Preparation of Example 1032
Preparation of Example 1033
Preparation of Example 1034
Preparation of Example 1035
Preparation of Example 1036
Preparation of Example 1037
Preparation of Example 1038
Preparation of Example 1039
The following examples were prepared following similar procedures described for the preparation for the aforementioned examples and general synthetic and purification procedures.
The following compounds were prepared following general procedures and general synthetic and purification procedures.
Method
LAG-3 cell binding assay: Human Raji cells expressing endogenous MHC Class II molecules were used for binding to either human LAG-3-mFc, mouse LAG-3, or cyno LAG-3-hFc proteins. Briefly Raji cells were plated in a 384-well plate (Coming 354663) at a density7 of 8000 cells/well. After 2 hour incubation at a 37 °C and 5% CO2 incubator, LAG-3 antigen (hLAG-3 -mFc, mLAG-3-mFc, or cLAG-3-hFc) were added to all wells at a final concentration of 0.088. 0.25. or 0.072 pg/ml and incubated for 30 minutes. Then, a corresponding detection antibody (R-Phycoerythrin conjugated anti-Mouse IgG, or anti-human IgG), Jackson Immuno Research Lab, PA) was added. The binding affinity7 of the LAG-3 antigen was quantified by reading the plate on a NXT High Content Reader (ThermoFisher). To assess the potency of LAG-3 compounds to block the binding of LAG-3 antigen to the MHCII molecules expressed on the Raji cell surface, compounds were serially diluted and added to the Raji cells prior to the addition of an appropriate LAG3 antigen. A: IC50 < 0.01 pM, B: 0.01 pM <= IC50 < 0.1 pM; C: 0.1 pM <= IC50 < 1 pM.
Table 1
Biological Data
Claims
1. A compound selected from
Example 1001, Example 1002. Example 1003, Example 1004, Example 1005, Example 1006, Example 1007, Example 1008, Example 1009, Example 1010, Example 1011, Example 1012, Example 1013, Example 1014, Example 1015, Example 1016, Example 1017, Example 1018, Example 1019, Example 1020, Example 1021, Example 1022, Example 1023, Example 1024, Example 1025. Example 1026, Example 1027, Example 1028, Example 1029, Example 1030. Example 1031. Example 1032. Example 1033, Example 1034, Example 1035, Example 1036, Example 1037, Example 1038 and Example 1039, Example 1040, Example 1041, Example 1042, Example 1043, Example 1044, Example 1045, Example 1046, Example 1047. Example 1048, Example 1049, Example 1050, Example 1051, Example 1052. Example 1053. Example 1054, Example 1055, Example 1056, Example 1057, Example 1058, Example 1059, Example 1060, Example 1061, Example 1062, Example 1063, Example 1064, Example 1065, Example 1066, Example 1067, Example 1068, Example 1069, Example 1070, Example 1071, Example 1072, Example 1073, Example 1074, Example 1075. Example 1076, Example 1077, Example 1078, Example 1079, Example 1080, Example 1081, Example 1082, Example 1083, Example 1084, Example 1085, Example 1086, Example 1087, Example 1088, Example 1089, Example 1090, Example 1091, Example 1092, Example 1093, Example 1094, Example 1095, Example 1096, Example 1097, Example 1098, Example 1099, Example 1100. Example 1101. Example 1102. Example 1103, Example 1104, Example 1105, Example 1106, Example 1107, Example 1108, Example 1109, Example 1110, Example 1111, Example 1112, Example 1113, Example 1114, Example 1115, Example 1116, Example 1117, Example 1118, Example 1119, Example 1120. Example 1121, Example 1122, Example 1123. Example 1124. Example 1125, Example 1126, Example 1127, Example 1128, Example 1129, Example 1130, Example 1131, Example 1132, Example 1133, Example 1134, Example 1135, Example 1136, Example 1137, Example 1138, Example 1139, Example 1140, Example 1141, Example 1142, Example 1143, Example 1144, Example 1145, Example 1146. Example 1147, Example 1148, Example 1149, Example 1150, Example 1151. Example 1152. Example 1153. Example
1154, Example 1155, Example 1156, Example 1157, Example 1158, Example 1159, Example 1160, Example 1161, Example 1162, Example 1163. Example 1164, Example 1165, Example 1166, Example 1167, Example 1168, Example 1 169, Example 1 170, Example 1171, Example 1172, Example 1173, Example 1174, Example 1175, Example 1176, Example 1177, Example 1178, Example 1179, Example 1180, Example 1181, Example 1182, Example 1183, Example 1184, Example 1185, Example 1186, Example 1187, Example 1188. Example 1189. Example 1190. Example 1191, Example 1192, Example 1193, Example 1194, Example 1195, Example 1196, Example 1197, Example 1198, Example 1199, Example 1200, Example 1201, Example 1202, Example 1203, Example 1204, Example 1205, Example 1206, Example 1207, Example 1208, Example 1209, Example 1210, Example 1211. Example 1212. Example 1213, Example 1214, Example 1215, Example 1216, Example 1217, Example 1218, Example 1219, Example 1220, Example 1221, Example 1222, Example 1223, Example 1224, Example 1225, Example 1226, Example 1227, Example 1228, Example 1229, Example 1230. Example 1231, Example 1232, Example 1233, Example 1234. Example 1235, Example 1236, Example 1237, Example 1238, Example 1239. Example 1240. Example 1241. Example 1242, Example 1243, Example 1244, Example 1245, Example 1246, Example 1247, Example 1248, Example 1249, Example 1250, Example 1251, Example 1252, Example 1253, Example 1254, Example 1255, Example 1256. Example 1257, Example 1258, Example 1259, Example 1260, Example 1261. Example 1262. Example 1263, Example 1264, Example 1265, Example 1266, Example 1267, Example 1268, Example 1269, Example 1270, Example 1271, Example 1272, Example 1273, Example 1274, Example 1275, Example 1276, Example 1277, Example 1278, Example 1279, Example 1280, Example 1281, Example 1282, Example 1283, Example 1284. or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 wherein the compound shows activity less than or equal to 1 pM in the LAG-3 cell binding assay.
3. The compound or a pharmaceutically acceptable salt thereof according to claim 2 as shown in the following tables:
Table 1
Table 2
Table 3
Table 4
Table 5
Table 7
4. A pharmaceutical composition comprising one or more compounds according to claim 1 in a pharmaceutically acceptable carrier.
5. A pharmaceutical composition comprising one or more compounds according to claim 2 in a pharmaceutically acceptable carrier.
6. A pharmaceutical composition comprising one or more compounds according to claim 3 in a pharmaceutically acceptable carrier.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202463563642P | 2024-03-11 | 2024-03-11 | |
| US63/563,642 | 2024-03-11 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025193572A1 true WO2025193572A1 (en) | 2025-09-18 |
Family
ID=95201071
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2025/019110 Pending WO2025193572A1 (en) | 2024-03-11 | 2025-03-10 | Macrocyclic peptides useful as immunomodulators |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025193572A1 (en) |
Citations (9)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2014008218A1 (en) | 2012-07-02 | 2014-01-09 | Bristol-Myers Squibb Company | Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof |
| WO2015042246A1 (en) | 2013-09-20 | 2015-03-26 | Bristol-Myers Squibb Company | Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors |
| AU2015364573A1 (en) * | 2014-12-19 | 2017-08-03 | Bristol-Myers Squibb Company | Immunomodulators |
| CN105504018B (en) * | 2016-01-13 | 2019-01-04 | 郑州大学 | LAG-3 affinity peptide N13, preparation method and applications |
| WO2020237050A1 (en) * | 2019-05-22 | 2020-11-26 | Leidos, Inc. | Lag3 binding peptides |
| CN110317245B (en) * | 2019-08-02 | 2021-04-09 | 郑州大学 | LAG-3 protein affinity cyclic peptide and its application |
| CA2905453C (en) * | 2013-03-15 | 2021-10-12 | Bristol-Myers Squibb Company | Macrocyclic inhibitors of the pd-1/pd-l1 and cd80(b7-1)/pd-l1 protein/protein interactions |
| CN116444616A (en) * | 2023-04-10 | 2023-07-18 | 郑州大学 | Affinity peptide, coupling peptide, medicament and application of LAG-3 protein |
| WO2023225661A1 (en) | 2022-05-20 | 2023-11-23 | Bristol-Myers Squibb Company | Macrocyclic immunomodulators |
-
2025
- 2025-03-10 WO PCT/US2025/019110 patent/WO2025193572A1/en active Pending
Patent Citations (9)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2014008218A1 (en) | 2012-07-02 | 2014-01-09 | Bristol-Myers Squibb Company | Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof |
| CA2905453C (en) * | 2013-03-15 | 2021-10-12 | Bristol-Myers Squibb Company | Macrocyclic inhibitors of the pd-1/pd-l1 and cd80(b7-1)/pd-l1 protein/protein interactions |
| WO2015042246A1 (en) | 2013-09-20 | 2015-03-26 | Bristol-Myers Squibb Company | Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors |
| AU2015364573A1 (en) * | 2014-12-19 | 2017-08-03 | Bristol-Myers Squibb Company | Immunomodulators |
| CN105504018B (en) * | 2016-01-13 | 2019-01-04 | 郑州大学 | LAG-3 affinity peptide N13, preparation method and applications |
| WO2020237050A1 (en) * | 2019-05-22 | 2020-11-26 | Leidos, Inc. | Lag3 binding peptides |
| CN110317245B (en) * | 2019-08-02 | 2021-04-09 | 郑州大学 | LAG-3 protein affinity cyclic peptide and its application |
| WO2023225661A1 (en) | 2022-05-20 | 2023-11-23 | Bristol-Myers Squibb Company | Macrocyclic immunomodulators |
| CN116444616A (en) * | 2023-04-10 | 2023-07-18 | 郑州大学 | Affinity peptide, coupling peptide, medicament and application of LAG-3 protein |
Non-Patent Citations (40)
| Title |
|---|
| "Fmoc Solid Phase Synthesis", 2000, OXFORD UNIVERSITY PRESS |
| "GENBANK", Database accession no. NP_054862 |
| ANSELL SM ET AL., AM J HEMATOL, vol. 60, 1999, pages 99 |
| ATHERTON, E ET AL.: "Special Methods in Peptide Synthesis, Part C", vol. 9, 1987, ACADEMIC PRESS, article "The Fluorenylmethoxycarbonyl Amino Protecting Group", pages: 1 - 38 |
| ATHERTON, E.SHEPPARD, R. C: "Solid Phase Peptide Synthesis: A Practical Approach", 1989, IRL PRESS |
| BALZANO, INT. J. CANCER, vol. 7, 1992, pages 28 - 32 |
| BARANY, G ET AL.: "Special Methods in Peptide Synthesis, Part A", vol. 2, 1980, ACADEMIC PRESS, article "The Peptides: Analysis, Synthesis, Biology", pages: 3 - 284 |
| BERRIEN-ELLIOTT, M ET AL., CANCER RESEARCH, vol. 73, no. 2, 2013, pages 605 - 616 |
| BLACKBUM SD ET AL., NAT. IMMUNOL, vol. 10, 2009, pages 29 - 37 |
| BLACKBUM SD, NAT. TMMUNOK, vol. 10, 2009, pages 29 - 37 |
| CAMISASCHI C ET AL., J. TMMUNOK, vol. 184, 2010, pages 6545 - 6551 |
| CASTELLI, ONCOIMMUNOLOGY, vol. 3, 2014, pages 11 |
| CODING, S. R. ET AL., JOURNAL OF IMMUNOLOGY, vol. 190, no. 9, 1950, pages 4899 - 909 |
| DOLCETTI R ET AL., INFECTIOUS AGENTS AND CANCER, vol. 5, 2010, pages 22 |
| GREEN MR ET AL., CLIN CANCER RES, vol. 18, 2012, pages 1611 |
| GROSSO JF, J. CLIN. INVEST, vol. 117, 2007, pages 3383 - 3392 |
| HUANG CT ET AL., IMMUNITY, vol. 21, 2004, pages 503 - 513 |
| HUARD, EUR. J. IMMUNOL, vol. 26, 1996, pages 1180 - 6 |
| HUARD, EUR. J. IMMUNOL., vol. 24, 1994, pages 3216 - 21 |
| HUARD, PROC. NATL. ACAD. SCI, vol. 94, 1997, pages 5744 - 9 |
| HUARD, PROC. NATL. ACAD. SCI., vol. 94, 1997, pages 5744 - 9 |
| KANAKRY JA ET AL., BLOOD, vol. 121, 2013, pages 3547 |
| KIESLOW, EUR. J. IMMUNOL, vol. 35, 2005, pages 2081 - 88 |
| KING, D.S ET AL., INT. J. PEPTIDE PROTEIN RES., vol. 36, 1990, pages 255 - 266 |
| KOTASKOVA J ET AL., J MOL DIAGN, vol. 12, no. 3, 2010, pages 328 - 334 |
| KOUO, CANCER IMMUNOL RES, vol. 3, no. 4, 2015, pages 412 - 23 |
| MEYERS E ET AL., COMPUT. APPL. BIOSCI., vol. 4, 1988, pages 11 - 17 |
| MONTI S ET AL., BLOOD, vol. 105, 2005, pages 1851 |
| MURATA, AM. J. PATHOL., vol. 155, 1999, pages 453 - 460 |
| NEEDLEMAN E, J. MOL. BIOL., vol. 48, 1970, pages 444 - 453 |
| STEWART, J. M. YOUNG, J.D: "Solid-Phase Peptide Synthesis", 1984, PIERCE CHEMICAL CO. |
| TRIEBEL F ET AL., J. EXP. MED., vol. 171, 1990, pages 1393 - 1405 |
| TSIMBERIDOU AM ET AL., LEUK LYMPHOMA, vol. 47, no. 2, 2006, pages 231 - 44 |
| WOO, S-R ET AL., CANCER RESEARCH, vol. 72, no. 4, 2011, pages 917 - 927 |
| WORKMAN CJ ET AL., J. IMMUNOL., vol. 182, no. 4, 2009, pages 1885 - 91 |
| WORKMAN CJ ET AL., J. TMMUNOK, vol. 174, 2005, pages 688 - 695 |
| WORKMAN, J. IMMUNOL, vol. 169, 2002, pages 5392 - 5 |
| XU, CANCER RES, vol. 74, no. 13, 2014, pages 3418 - 28 |
| ZHAI WENJIE ET AL: "A novel cyclic peptide targeting LAG-3 for cancer immunotherapy by activating antigen-specific CD8+ T cell responses", ACTA PHARMACEUTICA SINICA B, vol. 10, no. 6, 1 June 2020 (2020-06-01), pages 1047 - 1060, XP093077457, ISSN: 2211-3835, DOI: 10.1016/j.apsb.2020.01.005 * |
| ZHANG J ET AL., BMC BIOINFORMATICS, vol. 1, no. 1, 2010, pages S5 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| AU2014405919B2 (en) | Macrocyclic inhibitors of the PD-1/PD-L1 and CD80 (B7-1)/PD-Li protein/protein interactions | |
| US9879046B2 (en) | Macrocyclic inhibitors of the PD-1/PD-L1 and CD80(B7-1)/PD-L1 protein/protein interactions | |
| EP2160221B1 (en) | Cyclic peptide cxcr4 antagonists | |
| CN1756763B (en) | Dimerized peptides | |
| CN113811541A (en) | OX40 specific bicyclic peptide ligands | |
| IL223678A (en) | Immunosuppression modulating compounds | |
| JP2010500298A (en) | Peptides with pharmacological activity for the treatment of disorders associated with altered cell migration such as cancer | |
| WO2015022283A1 (en) | Yap-tead inhibitors | |
| US6780845B2 (en) | Compounds and methods for cancer therapy | |
| CA2405476A1 (en) | Compounds and methods for modulating endothelial cell adhesion | |
| US6207639B1 (en) | Compounds and methods for modulating neurite outgrowth | |
| EP4314012A1 (en) | Immunomodulators | |
| WO1995000546A1 (en) | Endothelin-antagonizing peptide | |
| WO2025193572A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| CN108640973B (en) | Polypeptide with targeting effect Syntenin protein, dimer thereof and application thereof | |
| WO2025193569A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| WO2025193574A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| WO2025193571A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| WO2025193573A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| WO2025193583A1 (en) | Macrocyclic peptides useful as immunomodulators | |
| CN108640974B (en) | A polypeptide targeting the PDZ domain of Syntenin protein and its dimer | |
| WO2025193770A1 (en) | Macrocyclic peptides | |
| WO2004031221A1 (en) | Tumor targeting agents and uses thereof | |
| JP2012180315A (en) | Cyclic peptide compound having protein tyrosine kinase inhibiting activity | |
| JP2014530825A (en) | Peptide compound derived from melanotransferrin and use thereof |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25715438 Country of ref document: EP Kind code of ref document: A1 |