The present invention relates to a class of antibody-drug conjugates of novel structural exatecan analogues. Specifically, the present disclosure relates to an antibody-drug conjugate of an exatecan analogue containing structural unit Y, a pharmaceutical composition comprising the conjugate and a use of the conjugate or pharmaceutical composition.
Antibody-drug conjugates (ADCs), as novel targeted drugs, generally consist of three components: an antibody or antibody-like ligand, a small molecule drug, and a linker that couples the ligand to the drug. ADCs utilize the specific recognition of antigens by antibodies to transport the drug molecules near the target cells and effectively release the drug molecules for therapeutic purposes. In 2000, Mylotarg, an ADC drug from Pfizer, was first launched on the market, since then, the promising and challenging field of ADCs has been in the public's view. In recent years, the pharmaceutical market has ushered in a new round of ADC research and development boom, with a total of 13 ADC drugs currently on the market worldwide.
As an antitumor small molecule compound, known as a camptothecin derivative that exhibits antitumor effects by inhibiting DNA topoisomerase I, exatecan was developed by Daiichi Sankyo, and was advanced to phase III clinical trials in the early stages of its use as a stand-alone chemotherapeutic agent, with the main indications for bone, prostate, breast, and pancreatic cancers. Unlike irinotecan, which is currently in clinical use, exatecan does not require activation through the use of enzymes. In addition, compared with SN-38, which is the pharmacodynamic principal of irinotecan, and topotecan, which is also used in the clinic, exatecan has stronger inhibitory activity against topoisomerase I, and has stronger cytotoxicity against a wide range of cancer cells in vitro. Exatecan has not been successfully marketed as a stand-alone chemotherapeutic agent and it is hypothesized that this is related to its higher cellular activity, resulting in a narrow therapeutic window.
Daiichi Sankyo/AstraZeneca co-developed and commercialized antibody-coupled drug DS-8201a (trade name: Enhertu) in December 2019, which combines exatecan with hydroxyacetic acid to form an amide derivative and linkage to form an ADC, as a next-generation antibody-coupled drug, Enhertu is already showing potential to become a blockbuster.
It is necessary and urgent to explore and discover exatecan derivatives with better antitumor activity to improve the safety and efficacy of antitumor small molecule compounds in ADC drug applications, so as to obtain antitumor drugs with excellent efficacy.
The present invention provides a ligand-drug conjugate or a pharmaceutically acceptable salt thereof, wherein the ligand-drug conjugate comprises a structure shown by formula (-D0):
—Rc—Ar1—(Cy)m-(CRaRb)m—X—C(═O)—, —N((CRaRb)m CF3)—C(═O)—, —Rc—(CRaRb)m—C(═O)—Rc—Ar1-(Cy)m-(CRaRb)m—X—C(═O)—, or
wherein the C(═O)— end of Y is connected to —NH— in the structure of formula (-D0); preferably, Y is
O—Ar1-(Cy)m-(CRaRb)m—X—C(═O)—, —NH—Ar1-(Cy)m-(CRaRb)m—X—C(═O)—, —N((CRaRb)mCF3)—C(═O)—, —O—(CRaRb)m—C(═O)—NH—Ar1—(Cy)m-(CRaRb)m—X—C(═O)—, or
wherein the C(═O)— end of Y is connected to —NH— in the structure of formula (-D0);
The present invention provides a ligand-drug conjugate or a pharmaceutically acceptable salt thereof, wherein the ligand-drug conjugate comprises a structure shown by formula (-D):
In some embodiments of the present invention, there provide a ligand-drug conjugate or a pharmaceutically acceptable salt thereof, which is a ligand-drug conjugate shown by the general formula (Pc-L-D0) or the general formula (Pc-L-D), or a pharmaceutically acceptable salt thereof:
Wherein, Y is as described above, n is an integer or a decimal from 1 to 15; preferably, n is an integer or a decimal from 1 to 13; preferably, n is an integer or a decimal from 1 to 10; more preferably, n is an integer or a decimal from 3 to 8;
In some embodiments of the present invention, Rc at each occurrence is each independently NH, or O; Rd and Re at each occurrence are each independently C(Rm)2 or NRm; Rf at each occurrence is CRm; and when the ring formed by Rd, Re, and Rf is heterocyclyl, the heterocyclyl contains 1, 2, or 3 heteroatoms, and the heteroatoms are each independently selected from N and O;
In some embodiments, Rm at each occurrence is each independently H, halogen, —OH, —CN, —NO2, —CF3, —C1-6 alkyl, —OC1-6 alkyl, —NH2, —NH(C1-6 alkyl), or —N(C1-6 alkyl)2, preferably H, halogen, —OH, —CN, —CF3, —NO2, C1-6 alkyl, or —OC1-6 alkyl, preferably H, halogen, —OH, —CN, —CF3, —NO2, C1-3 alkyl, or —OC1-3 alkyl, preferably —Cl, —Br, —F, —OH, —CN, —CF3, —NO2, —CH3, or —OCH3;
In some embodiments, p1 is an integer of 0, 1, 2, or 3, p2 is an integer of 1, 2, 3, or 4, and 2≤p1+p2≤4;
In some embodiments, the Cy at each occurrence is each independently selected from 6-10 membered arylene, or 5-10 membered heteroarylene, preferably selected from phenylene, or 5-6 membered heteroarylene; preferably, the heteroarylene contains 1, 2, or 3 heteroatoms, and the heteroatoms are independently selected from N, O, and S; the heteroarylene is unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-6 alkyl, —OC1-6 alkyl, —NH2, —NH(C1-6 alkyl), and —N(C1-6 alkyl)2, preferably unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-6 alkyl, and —OC1-6alkyl, preferably unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-3 alkyl, and —OC1-3 alkyl, preferably unsubstituted or optionally substituted with a substituent selected from —Cl, —Br, —F, —OH, —CN, —CH3, and —OCH3; In some embodiments, Ar1 at each occurrence is each independently selected from 6-10 membered arylene, preferably selected from phenylene; the arylene or phenylene is unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-6 alkyl, —OC1-6 alkyl, —NH2, —NH(C1-6 alkyl), and —N(C1-6 alkyl)2, preferably unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-6 alkyl, and —OC1-6 alkyl, preferably unsubstituted or optionally substituted with a substituent selected from halogen, —OH, —CN, —CF3, —NO2, C1-3 alkyl, and —OC1-3 alkyl, preferably unsubstituted or optionally substituted with a substituent selected from —Cl, —Br, —F, —OH, —CN, —CF3, —NO2, —CH3, and —OCH3;
In some embodiments, Ra and Rb are identical or different and are each independently selected from hydrogen atom, deuterium atom, halogen, C1-3 alkyl, halo C1-3 alkyl, deuterated C1-3alkyl, —OC1-3 alkyl, —OH, —NH2, —CN, —CF3, —NO2, and C1-3 alkylene-OH; preferably, Ra and Rb are identical or different and are each independently selected from hydrogen atom, deuterium atom, and halogen; and/or
In some embodiments, X is independently selected from single bond, —NH—, or O; m at each occurrence is each independently an integer of 0, 1, 2, or 3.
In some embodiments of the present invention, Y is selected from:
wherein p1 is an integer of 0, 1, 2, or 3, p2 is an integer of 1, 2, 3, or 4, and 2≤p1+p2≤4; X is independently selected from single bond, —NH—, or O, and n is an integer of 0, 1, 2, or 3;
In some embodiments of the present invention, Y is selected from:
In some embodiments of the present invention, the linker unit -L- is -L1-L2-L3-L4-, L1 is selected from -(succinimidyl-3-yl-N)—W—C(═O)—, wherein W is C1-10 alkylene, C1-10 alkylene-cycloalkylene, C1-10 heteroalkylene, C1-10 alkylene-heterocyclylene, or C1-10 heteroalkylene-cycloalkylene, preferably W is alkylene, alkylene-cycloalkylene, or C1-8 heteroalkylene, the heteroalkyl contains 1-3 heteroatoms each independently selected from N, O or S, wherein the C1-8alkyl, cycloalkyl and C1-8heteroalkylene are unsubstituted or each independently optionally further substituted with one or more substituents selected from halogen, hydroxyl, —CN, amino, C alkyl, haloalkyl, deuterated alkyl, alkoxy and cycloalkyl, preferably unsubstituted or each independently optionally further substituted with one or more substituents selected from halogen, hydroxyl, —CN, amino, C1-6 alkyl, haloC1-6alkyl, deuterated C1-6 alkyl, C1-6 alkoxy and C5-8 cycloalkyl, preferably unsubstituted or optionally substituted with one or more substituents selected from halogen, —OH, —CN, C1-3 alkyl, and —OC1-3 alkyl, preferably unsubstituted or optionally substituted with a substituent selected from Cl, Br, F, —OH, —CN, methyl, and —OCH3;
In some embodiments of the present invention, L1 is selected from -(succinimidyl-3-yl-N)—(CH2)s—C(═O)— and -(succinimidyl-3-yl-N)—CH2-cyclohexyl-C(═O)—, wherein s is an integer of 2, 3, 4, 5, 6, 7, or 8.
In some embodiments of the present invention, L2 is selected from —NR4(CH2CH2O)rCH2CH2C(═O)—, and —NR4CH2—Ar2—(CH2CH2O)rCH2CH2NR4C(═O)CH2OCH2C(═O)—, wherein r is an integer from 1 to 20; preferably r is an integer of 1, 2, 3, 4, 5, 6, 7, or 8, and Ar2 is
In some embodiments of the present invention, L3 is a peptide residue consisting of 2 to 7 amino acids selected from alanine, phenylalanine, glycine, valine, lysine, citrulline, serine, glutamic acid, and aspartic acid; preferably L3 is a dipeptide residue, tripeptide residue, or tetrapeptide residue consisting of amino acids selected from alanine, phenylalanine, glycine, valine, lysine, citrulline, serine, glutamic acid, and aspartic acid; more preferably L3 is selected form dipeptide residue of valine-alanine, tripeptide residue of alanine-alanine-alanine, or tetrapeptide residue of glycine-glycine-phenylalanine-glycine.
In some embodiments of the present invention, L4 is selected from —NR5(CR6R7)t—Z—(CR6R7)t—C(═O)— or —NR5—Ar3—(CR6R7)t—Z—C(═O)—, t at each occurrence is independently an integer of 1, 2 or 3;
In some embodiments of the present invention, the linker unit -L- is:
In some embodiments of the present invention, there provide a ligand-drug conjugate or the pharmaceutically acceptable salt thereof, selected from the following structural formulas:
In some embodiments of the present invention, Pc is an antibody or an antigen-binding fragment thereof or a polypeptide, wherein the antibody is selected from a chimeric antibody, a humanized antibody and a fully human antibody;
Another aspect of the present invention provides a compound shown by the general formula (E0) or formula (E), or a pharmaceutically acceptable salt thereof:
Preferably, the compound of formula (E0) or formula (E) is not
In some embodiments of the present invention, Y is selected from:
wherein p1 is an integer of 0, 1, 2, or 3, p2 is an integer of 1, 2, 3, or 4, and 2≤p1+p2≤4; X is independently selected from single bond, —NH— or O, and m is an integer of 0, 1, 2, or 3; or
In some embodiments of the present invention, Y is selected from:
The compounds of the present invention shown in the formula (E), including but not limited to:
Another aspect of the present invention provides a compound having the structure shown in the following formula (I):
L1-L2-L3-L4 (I)
In some embodiments of the present invention, the compound of formula (I) is selected from:
Another aspect of the present invention further relates to a pharmaceutical composition comprising a therapeutically effective amount of a ligand-drug conjugate or compound as described herein, or a pharmaceutically acceptable salt or solvent compound thereof, and one or more pharmaceutically acceptable carriers.
Another aspect of the present invention also relates to a pharmaceutical formulation comprising a compound of formula I or a pharmaceutically acceptable salt thereof as an active ingredient, or a pharmaceutical composition of the present invention. In some embodiments, the formulation is in the form of a solid formulation, a semi-solid formulation, a liquid formulation or a gaseous formulation.
The pharmaceutical compositions of the present invention may be administered by a variety of routes, depending on whether topical or systemic treatment is required and the area to be treated. The drug may be administered topically (e.g., transdermally, dermally, ocularly, and mucosally including intranasal, vaginal, and rectal delivery), pulmonary (e.g., by inhalation or blowing in a powder or aerosol, including through a nebulizer; intratracheal, intranasal), orally, or parenterally. Parenteral delivery includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intracerebroventricular delivery.
Another aspect of the present invention is to provide a product, for example in the form of a kit. The product as used herein is intended to include, but is not limited to, a kit and packaging. The kit may also include instructions for use.
Another aspect of the present invention further relates to ligand-drug conjugates or compounds, or pharmaceutically acceptable salts or solvent compounds thereof, or pharmaceutical compositions thereof, as described herein, which are used as drugs, Another aspect of the present invention further relates to the use of a ligand-drug conjugate or compound, or a pharmaceutically acceptable salt or solvates thereof, or a pharmaceutical composition thereof, as described herein in the preparation of a drug for the treatment or prevention of tumors; preferably, wherein the tumor is a cancer associated with TROP-2 expression, or the tumor is a cancer associated with HER3 expression; preferably, the HER3 expression-associated cancer is selected from non-small cell lung cancer, melanoma, breast cancer and colorectal adenocarcinoma; preferably, the TROP-2 expression-associated cancer are selected from breast cancer, triple negative breast cancer, gastric cancer and pancreatic cancer.
Another aspect of the present invention further relates to a method of treating and/or preventing a tumor, including administering a therapeutically effective amount of the ligand-drug conjugate or the pharmaceutically acceptable salt thereof, or the compound or the pharmaceutically acceptable salt thereof or the pharmaceutical composition as described herein to a subject in need thereof; preferably, wherein the tumor is a cancer associated with TROP-2 expression, or the tumor is a cancer associated with HER3 expression;
Another aspect of the present invention further relates to a ligand-drug conjugate or a pharmaceutically acceptable salt thereof, or a compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described herein, for use in the prevention or treatment of a tumor; preferably, wherein the tumor is a cancer associated with TROP-2 expression.
The exatecan analogues provided by the present invention have strong tumor inhibition efficiency, and the TROP-2 antibody-drug conjugates and HER-3 antibody-drug conjugates provided by the present invention have good tumor inhibition effect, which is suitable for clinical drug application.
Unless otherwise defined hereinafter, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by those skilled in the art.
Certain compounds of the present invention may be present in free form or, where appropriate, in the form of pharmaceutically acceptable derivatives thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, prodrugs, stereoisomers (including, but not limited to, diastereomers and enantiomers), tautomers, solvates, polymorphs and isotopic compounds, which are capable of delivering the compounds of the present invention or metabolites thereof, either directly or indirectly, upon administration to a patient in need thereof. Accordingly, when reference is made herein to “the compounds of the present invention”, it is also intended to cover the various derivative forms of the compounds described above.
The term “pharmaceutically acceptable salt” refers to a salt that retains the biological potency of the free acids and bases of a particular compound without biological adverse effects. Examples of pharmaceutically acceptable salts include, but are not limited to: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, sulfuric acid, hydrobromic acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as malic acid, fumaric acid, maleic acid, benzoic acid, phenylacetic acid, succinic acid, tartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, hydroxyacetic acid, cinnamic acid, pyruvic acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, acrylic acid, mandelic acid, and the like; or (2) alkali addition salts formed with alkali metals such as lithium, sodium, potassium, and the like; with alkaline earth metals such as calcium, magnesium, and the like; and with organic bases such as ammonium, choline, diethanolamine, lysine, ethylenediamine, tertiary-butylamine, tertiary-octylamine, tris(hydroxymethyl)aminomethane, N-methylglucosamine, triethanolamine, dehydroabietic amine, and the like. Other pharmaceutically acceptable salts are known to those skilled in the art.
Prodrugs of the compounds of the present invention are included within the scope of protection of the present invention. Typically, the prodrugs are functional derivatives that are readily converted in vivo to the desired compounds. Accordingly, the term “administer” in the therapeutic methods provided by the present invention encompasses administering a compound disclosed in the present invention, or, a compound, although not expressly disclosed in the present invention, that is capable of being converted in vivo to a compound disclosed in the present invention upon administration to a subject for the treatment of the various diseases described. Conventional methods for selecting and preparing suitable prodrugs derivatives are documented, for example, in the book Design of Prodrugs (H. Bundgaard, Elsevier, 1985).
The compounds described in the present invention may contain one or more asymmetric centers and may result in diastereomers and optical isomers. The present invention includes all possible diastereomerss and racemic mixtures thereof, substantially pure split enantiomers thereof, all possible geometric isomers and pharmaceutically acceptable salts thereof.
The compounds of the present invention do not precisely define the stereo structure of the compound at a particular position. The present invention includes all stereoisomers of the compounds and pharmaceutically acceptable salts thereof. Moreover, mixtures of stereoisomers and isolated specific stereoisomers are included in the present invention. The products produced during the synthetic process of preparing such compounds, or using racemization or differential isomerization methods well known to those of ordinary skill in the art, may be mixtures of stereoisomers.
When tautomers of the compounds of the present invention exist, the present invention includes any possible tautomers and pharmaceutically acceptable salt thereof, and mixtures thereof, unless specifically stated.
When the compounds of the invention and pharmaceutically acceptable salts thereof are present in the form of solvates or polymorphs, the invention encompasses any possible solvate and polymorph forms. The type of solvent used to form the solvates is not specifically limited, as long as the solvent is pharmaceutically acceptable. For example, water, ethanol, propanol, acetone, and similar solvents may be employed.
The present invention also includes all pharmaceutically acceptable isotope compounds which are identical to the compounds of the present invention except that one or more atoms are replaced by atoms having the same atomic number but a different atomic mass or mass number than that which predominates in nature. Examples of isotopes suitable for inclusion in the compounds of the present invention include, but are not limited to, isotopes of hydrogen (e.g., deuterium (2H), tritium (3H)); isotopes of carbon (e.g., 13C and 14C); isotopes of chlorine (e.g., 37Cl); isotopes of iodine (e.g., 125I); isotopes of nitrogen (e.g., 13N and 15N); isotopes of oxygen (e.g., 17O and 8O); isotopes of phosphorus (e.g., 32P); and isotopes of sulfur (e.g., 34S).
The term “ligand” is a macromolecular compound that recognizes and binds an antigen or receptor associated with a target cell. The role of the ligand is to present the drug to the target cell population to which the ligand is bound, in this embodiment of the invention the ligand is denoted as Pc, and the ligand may form a linkage with a linkage unit by means of a heteroatom on the ligand, preferably an antibody, or an antigen-binding fragment thereof, or a polypeptide, and the antibody is selected from chimeric antibodies, humanized antibodies, fully human or murine antibodies; preferably a monoclonal antibody.
The term “drug” refers to a cytotoxic drug, denoted as E or E0, which is a chemical molecule that has a strong ability to disrupt the normal growth of tumor cells.
The term “linker unit” or “linkage fragment” or “linkage unit” refers to a fragment or bond of a chemical structure that is attached to a ligand at one end and to the drug at the other end, or can be attached to other linkers and then connected to the drug. A preferred embodiment of the present disclosure is represented by L and L1 to L4, wherein the L1 end is attached to the ligand and the L4 end is attached to the structural unit Y and then to the drug (E or E0).
The term “ligand-drug conjugate” refers to the attachment of a ligand to a biologically active drug through a stabilized linkage unit. In the present disclosure “ligand-drug conjugate” is preferably an antibody-drug conjugate (ADC), which refers to the attachment of a monoclonal antibody or antibody fragment to a biologically active toxic drug through a stabilized linkage unit.
The three-letter codes and single-letter codes for amino acids used in this disclosure are as described in J. biol. chem, 243, p3558 (1968).
The term “antibody” refers to immunoglobulins, which are tetrapeptide chains consisting of two identical heavy chains and two identical light chains linked by interchain disulfide bonds. Immunoglobulins differ in the composition and order of amino acids in the constant region of the heavy chain, and therefore differ in their antigenicity. Accordingly, immunoglobulins can be categorized into five classes, or isoforms of immunoglobulins, i.e., IgM, IgD, IgG, IgA, and IgE, and the corresponding heavy chains are μ-chain, δ-chain, γ-chain, α-chain, and ε-chain thereof, respectively. The same class of Ig can be divided into different subclasses according to the differences in the amino acid composition of its hinge region and the number and position of disulfide bonds in the heavy chain, e.g., IgG can be classified into IgG1, IgG2, IgG3 and IgG4. light chain is classified into κ-chain or λ-chain by the differences in the constant region. Each of the five classes of Ig may have either a κ chain or a λ chain. The antibodies described in the present disclosure are preferably specific antibodies against cell surface antigens on target cells, and non-limiting embodiments are the following antibodies: anti-TROP-2 antibody, anti-HER2 (ErbB2) antibody, anti-EGFR antibody, anti-B7-H3 antibody, anti-c-Met antibody, anti-HER3 (ErbB3) antibody, anti-HER4 (ErbB4) antibody, anti-LIV-1 antibody, anti-ROR1 antibody, anti-CD20 antibody, anti-CD22 antibody, anti-CD30 antibody, anti-CD33 antibody, anti-CD44 antibody, anti-CD56 antibody, anti-CD70 antibody, anti-CD73 antibody, anti-CD105 antibody, anti-CEA antibody, anti-A33 antibody, anti-Cripto antibody, anti-EphA2 antibody, anti-G250 antibody, anti-MUC1 antibody, anti-Lewis Y antibody, anti-VEGFR antibody, anti-GPNNMB antibody, anti-Integrin antibody, anti-PSMA antibody, anti-Tenascin-C antibody, anti-SLC44A4 antibody, anti-Mesothelin antibody, or antigen-binding fragments thereof; more preferably, the antibody or antigen-binding fragment thereof is an anti-TROP-2 antibody, an anti-HER2 (ErbB2) antibody, anti-HER3 (ErbB3) antibody, anti-LIV-1 antibody, anti-ROR1 antibody, or antigen-binding fragments thereof; most preferably, the antibody or antigen-binding fragment thereof is sacituzumab, trastuzumab, or pertuzumab.
The antibodies of the present invention include murine antibodies, chimeric antibodies, humanized antibodies and fully human antibodies, preferably humanized antibodies and fully human antibodies.
The term “murine antibody” is used in the present invention to mean an antibody prepared from a mouse according to the knowledge and skill in the art. The preparation involves injecting to a test subject with a specific antigen and then isolating a hybridoma expressing an antibody having the desired sequence or functional properties.
The term “chimeric antibody” is used to describe an antibody made by fusing the variable region of a murine antibody with the constant region of a human antibody, which can reduce the immune response induced by the murine antibody. To create a chimeric antibody, a hybridoma secreting a murine-specific monoclonal antibody is established, the variable region gene is cloned from the murine hybridoma cells, the constant region gene of the human antibody is cloned according to the need, and the chimeric gene is inserted into an expression vector by linking the murine variable region gene and the human constant region gene, and then the chimeric antibody molecule is expressed in a eukaryotic system or a prokaryotic system.
The term “humanized antibody”, also known as CDR-grafted antibody, refers to antibodies produced by transplanting murine CDR sequences into the variable region framework of human antibodies, i.e., the framework sequences of different types of human germline antibodies. It can overcome the heterologous reaction induced by chimeric antibodies due to carrying a large number of murine protein components.
The term “fully human antibody”, also known as “fully human monoclonal antibodies”, is used to describe antibodies that are of human origin in both the variable and constant regions, removing immunogenicity and toxicity.
The term “antigen-binding fragment” refers to one or more fragments of an antibody that maintain the ability to specifically bind antigen. It has been shown that fragments of a full-length antibody can be utilized for the antigen-binding function of the antibody. Examples of binding fragments included in “antigen-binding fragment” include (i) a Fab fragment, a monovalent fragment comprising the structural domains of VL, VH, CL, and CH1, (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge over a hinge region, (iii) a F′d fragment comprising the structural domains of VH and CH1, (iv) an Fv fragment comprising the VH and VL structural domains of the single arm of the antibody, (v) a single structural domain or dAb fragment (Ward et al, (1989) Nature 341:544-546) consisting of the VH structural domain, and (vi) an isolated complementarity determining region (CDR) or (vii) a combination of two or more isolated CDRs optionally connected by a synthesized linker. In addition, although the two structural domains VL and VH of the Fv fragment are encoded by separate genes, recombinant methods may be used to connect them by synthetic linker, thereby enabling them to produce a single protein chain for which the VL and VH regions pair to form a monovalent molecule (referred to as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci USA 85:5879-5883). Such single-chain antibodies are also intended to be included in the term “antigen-binding fragment” of an antibody. Such antibody fragments are obtained using conventional techniques known to those skilled in the art, and the fragments are screened for utility in the same manner as for intact antibodies. The antigen-binding portion may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact immunoglobulins.
Fab is an antibody fragment having a molecular weight of about 50,000 and antigen-binding activity from a fragment obtained by treatment of an IgG antibody molecule with the protease papain (cleaving amino acid residue at position 224 of the H-chain), wherein about one-half of the N-terminal side of the H-chain and the whole of the L-chain are bound together by disulfide bonds.
F(ab′)2 is an antibody fragment having a molecular weight of about 100,000 and antigen-binding activity and containing two Fab regions linked at the hinge position, obtained by digesting the lower portion of the two disulfide bonds in the hinge region of IgG with the enzyme pepsin.
Fab′ is an antibody fragment having a molecular weight of about 50,000 and antigen-binding activity obtained by cleaving a disulfide bond in the hinge region of F(ab′)2 described above.
In addition, the Fab′ can be produced by inserting DNA encoding a Fab′ fragment of an antibody into a prokaryotic expression vector or a eukaryotic expression vector and introducing the vector into a prokaryotic or eukaryotic organism to express Fab′.
The terms “single-chain antibody”, “single-chain Fv” or “scFv” mean a molecule comprising an antibody heavy chain variable structure domain (or region; VH) and an antibody light chain variable structure domain (or region; VL) linked by a linker. Such scFv molecules may have the general structure: NH2—VL-linker-VH—COOH or NH2—VH-linker-VL-COOH. Suitable prior art linkers comprise repeated GGGGS amino acid sequences or variants thereof, e.g., using variants of 1-4 repeats (Holliger et al. (1993), Proc. Natl. Acad. Sci. USA 90:6444-6448). Other linkers that can be used in the present disclosure are as described by Alfthan et al. (1995), Protein Eng. 8:725-731, Choi et al. (2001), Eur. J. Immuno 1. 31:94-106, Hu et al. (1996), Cancer Res. 56:3055-3061, Kipriyanov et al. (1999), J. Mol. Biol. 293:41-56 and Roovers et al. (2001), Cancer Immunol.
The term “CDR” refers to one of the six highly variable regions within the variable structural domain of an antibody that primarily contribute to antigen binding. One of the most commonly used definitions of the 6 CDRs is provided by Kabat E. A. et al, (1991) Sequences of proteins of immunological interest. (NIH Publication 91-3242). As used herein, the Kabat definition of CDR should only be applied to CDR1, CDR2, and CDR3 (CDR L1, CDR L2, CDR L3 or L1, L2, L3) of light chain variable structural domains, and CDR2 and CDR3 (CDR H2, CDR H3 or H2, 113) of heavy chain variable structural domains.
The term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been attached. In one embodiment, the vector is a “plasmid”, which refers to a circular double-stranded DNA loop to which an additional DNA segments can be ligated. In another embodiment, the vector is a viral vector in which additional DNA segments can be attached to a viral genome. The vectors disclosed herein are capable of replicating autonomously in a host cell into which they have been introduced (e.g., bacterial vectors and additive mammalian vectors having the replication start point of a bacterium) or may be integrated into the genome of the host cell after introduction into the host cell, thereby replicating along with the host genome (e.g., non-episomal mammalian vectors).
The term “alkyl” refers to a saturated aliphatic hydrocarbon group, which is a straight or branched chain group comprising 1 to 20 carbon atoms, preferably an alkyl comprising 1 to 12 carbon atoms, more preferably an alkyl comprising 1 to 10 carbon atoms, and most preferably an alkyl comprising 1 to 6 carbon atoms. The term “C1-6 alkyl” refers to a saturated straight or branched hydrocarbon group having 1 to 6 carbon atoms (e.g. 1, 2, 3, 4, 5 or 6 carbon atoms). For example, “C1-6 alkyl” may be methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, or n-hexyl.
The term “cycloalkyl” refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, wherein the cycloalkyl ring comprises 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 10 carbon atoms, and most preferably 3 to 8 carbon atoms. Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyls include spiro, fused and bridged cycloalkyls.
The term “heterocyclyl” refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising from 3 to 20 ring atoms, wherein one or more (e.g., 1, 2, or 3) of the ring atoms are selected from nitrogen, oxygen, or sulfur and the remaining ring atoms are carbon. Preferably, it contains from 3 to 12 ring atoms, wherein 1 to 4 of which are heteroatoms; more preferably the cycloalkyl ring contains from 3 to 10 ring atoms. Non-limiting examples of monocyclic heterocyclyls include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. Polycyclic heterocyclyls include spiro, fused and bridged heterocyclyls.
The term “aryl” refers to 6 to 14 membered all-carbon monocyclic or densely fused polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) groups having a conjugated π-electronic system, preferably 6 to 10 membered, e.g., phenyl and naphthyl, preferably phenyl. The aryl ring may be densely bonded to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring.
The term “heteroaryl” refers to a heteroaromatic system comprising 1 to 4 heteroatoms and 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl groups are preferably from 5 to 10 membered, more preferably 5 or 6 membered, such as furanyl, thiophenyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidyl, pyrazinyl, imidazolyl, tetrazolyl and the like. The heteroaryl ring may be fused on an aryl, heterocyclic or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring.
The term “haloalkyl” means an alkyl substituted with one or more halogens.
The term “deuterated alkyl” means an alkyl substituted with one or more deuterium atoms.
The term “halogen” means fluorine, chlorine, bromine or iodine.
The terms “substitute” and “substituted” mean that one or more (e.g., one, two, three, or four) hydrogens on the designated atom are substituted with a selection from the indicated moiety, provided that the normal atomic valence of the designated atom is not exceeded in the present context, and the substitution forms a stable compound. Combinations of substituents and/or variables are permitted only if such combinations form stable compounds.
If the substituent is described as “optionally . . . substituted”, the substituent may be (1) unsubstituted or (2) substituted. If an atom or group is described as being optionally substituted with one or more of a list of substituents, one or more hydrogens on the atom or group may be replaced with an independently selected, optional substituent. If the substituents are described as “independently selected from” or “each independently are”, the substituents are each selected independently. Thus, each substituent may be the same as or different from another (other) substituent. For example, when a substituent or substituent position or a different substituent or substituent position has an R group (e.g., but not limited to, R2, R3, Rh, Ri, Rx and/or Ry) that may have identical or different symbols, the individual R are each selected independently, i.e., they may be identical or different. The same applies to the selection of numerical values such as d, g, m, and n.
Unless indicated, as used herein, the linkages of substituent may be derived from any suitable position of the substituent.
When the bond of the substituent is shown to be through a bond connecting two atoms in the ring, then such a substituent may be bonded to any of the ring-forming atoms in the substituted ring.
The terms “include”, “comprise”, “have”, “contain” or “involve” and other variations thereof are inclusive or open-ended herein and do not exclude other elements or method steps not enumerated. It should be understood by those skilled in the art that terms such as “comprising” cover the meaning of “consisting of”.
In the present invention, “pharmaceutically acceptable carrier” means a diluent, excipient, or medium that is administered with the active ingredient and that is suitable, within the bounds of reasonable medical judgment, for exposure to the tissues of human beings and/or other animals without undue toxicity, irritation, allergic reaction, or other problems or complications commensurate with a reasonable benefit/risk ratio. The term “active ingredient”, “therapeutic agent”, “active substance” or “active agent” refers to a chemical entity that is effective in treating one or more symptoms of a target disease or condition.
As used herein, the term “effective amount” (e.g., “therapeutically effective amount” or “preventively effective amount”) refers to an amount of an active ingredient that, when administered, achieves the desired effect to a certain extent, such as relieving one or more symptoms of the condition being treated or preventing the occurrence of the condition or its symptoms.
Unless otherwise indicated, as used herein, the term “treating” means reversing, mitigating, inhibiting the progression of, the disease or condition which to the term applies, or one or more symptoms of the disease or condition, or preventing the disease or condition, or one or more symptoms of the disease or condition.
The experimental methods in the following embodiments are conventional methods unless otherwise specified. The chemical raw materials, reagents and the like used in the following embodiments are commercially purchased products, unless otherwise specified. Abbreviations and their meanings appearing herein are shown below:
Unless otherwise stated, the cell strains or cell lines used in the embodiments of the present invention are available commercially. Wherein,
EXD (25 mg, 47 μmol, 1.0 eq, purchased from MedChemExpress, item no. HY-13631A) and compound 1a (9.5 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were add and stirred for 2 h at room temperature. Water was added, extracted with ethyl acetate, and the organic phase was washed with saturated saline, dried, concentrated and then purified by column chromatography (9% MeOH in CH2Cl2) to give a white solid (29 mg, 100%). MS (ESI): m/z found [M−99]+=519.2.
Compound 1b (28 mg, 45 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 3 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (32% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (28.6 mg, 100%). MS (ESI): m/z found [M+1]+=519.2.
1H NMR (400 MHz, DMSO-d6): δ 8.76 (br, 2H), 8.69 (d, J=8.4 Hz, 1H), 7.82 (d, J=11.2 Hz, 1H), 7.32 (s, 1H), 6.52 (br, 1H), 5.62 (t, J=4.0 Hz, 1H), 5.43 (s, 2H), 5.23 (q, J=19.8 Hz, 2H), 4.10-4.02 (m, 4H), 3.18-3.12 (m, 2H), 2.41 (s, 3H), 2.19-2.16 (m 2H), 0.87 (t, J=7.2 Hz, 3H).
p-Aminophenylacetic acid 2a (617 mg, 4.08 mmol, 1.0 eq) was dissolved in dioxane (8 mL) and water (4 mL), and 1N sodium bicarbonate (4 mL) and (Boc)2O (980 mg, 4.49 mmol, 1.1 eq) were added, and stirred for 24 h at room temperature. The reaction solution was neutralized with 2N hydrochloric acid, diluted with 50 mL of water, extracted with ethyl acetate, washed with saturated brine, dried and concentrated and purified by column chromatography (25% EA in PE) to obtain a white solid (700 mg, 68%), MS (ESI): m/z found [M−1]−=250.0.
EXD (25 mg, 47 gmol, 1.0 eq) and compound 2a (11.8 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. Water was added, extracted with ethyl acetate, and the organic phase was washed with saturated saline, dried and concentrated and purified by column chromatography (4% MeOH in CH2Cl2) to obtain a white solid (27 mg, 86%), MS (ESI): m/z found [M+1]+=669.2.
Compound 2c (27 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and purified by reversed-phase column chromatography (310% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (19.3 mg, 70%), MS (ESI): m/z found [M+1]+=569.2.
1H NMR (400 MHz, DMSO-d6): δ 8.71 (d, J=8.4 Hz, 1H), 7.82 (d, J=10.8 Hz, 1H), 7.32-7.30 (m, 3H), 7.11 (d, J=8.0 Hz, 2H), 6.55 (br, 1H), 5.56-5.50 (m, 1H), 5.44 (s, 2H), 5.21 (dd, J=18.4, 32.8 Hz, 2H), 3.50-3.18 (m, 4H), 2.42 (s, 3H), 2.14-2.12 (m, 2H), 1.93-1.82 (m, 2H), 0.88 (t, J=7.2 Hz, 3H).
Hydroxyacetic acid (2.0 mg, 26 μmol, 1.0 eq) was dissolved in DMF (1 mL). EDCI·HCl (6.1 mg, 32 μmol, 1.2 eq) and HOSu (3.7 mg, 32 μmol, 1.2 eq) were added, and stirred for 2 h at room temperature. Compound 2 (18 mg, 26 μmol, 1.0 eq) and DIEA (11 μL, 66 μmol, 2.5 eq) were added and stirring was continued for 10 hours. The reaction solution was purified by HPLC to obtain a pale yellow solid (10.0 mg, 61%), MS (ESI): m/z found [M+1]+=627.2.
1H NMR (400 MHz, DMSO-d6): δ 9.59 (s, 1H), 8.66 (t, J=9.0 Hz, 1H), 7.79 (d, J=11.0 Hz, 1H), 7.59 (d, J=8.0 Hz, 2H), 7.29 (d, J=1.9 Hz, 1H), 7.20 (dd, J=8.4, 3.5 Hz 2H), 6.52 (s, 1H), 5.65 (s, 1H), 5.51 (dt, J=9.1, 4.9 Hz, 1H), 5.42 (d, J=3.3 Hz, 2H), 5.24 (dd, J=18.9, 2.9 Hz, 1H), 5.12 (dd, J=19.0, 4.6 Hz, 1H), 3.95 (d, J=4.2 Hz, 2H), 3.43 (d, J=4.0 Hz, 2H), 3.16 (d, J=6.0 Hz, 2H), 2.39 (s, 3H), 2.15-2.06 (m, 2H), 1.90-1.79 (m, 2H), 0.85 (td, J=7.4, 2.8 Hz, 3H).
EXD (25 mg, 47 μLmol, 1.0 eq) and N-Boc-L-proline 4a (10 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL), and HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution changed from turbid to clear. Water was added, and the reaction solution was extracted with ethyl acetate, washed with saturated saline, dried, concentrated and purified by column chromatography (4% MeOH in CH2C2) to obtain a white solid (26 mg, 88%), MS (ESI): m/z found [M+1]+=633.2.
Compound 4b (27 mg, 43 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and purified by reversed-phase column chromatography (31% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (18.5 mg, 67%), MS (ESI): m/z found [M+1]+=533.2.
1H NMR (400 MHz, DMSO-d6): δ 9.11 (d, J=8.4 Hz, 1H), 7.84 (d, J=10.8 Hz, 1H), 7.32 (s, 1H), 6.55 (s, 1H), 5.65-5.61 (m, 1H), 5.43 (s, 2H), 5.30 (d, J=18.8 Hz, 1H), 5.09 (d, J=18.8 Hz, 1H), 4.17 (s, 1H), 3.38-3.21 (m, 4H), 2.42 (s, 3H), 2.25-2.20 (m, 3H), 1.94-1.83 (m, 5H), 0.88 (t, J=7.2 Hz, 3H).
Hydroxyacetic acid (3.5 mg, 46 μmol, 1.0 eq) was dissolved in DMF (1 mL). EDCI-HCl (10.7 mg, 56 μmol, 1.2 eq) and HOSu (6.4 mg, 56 μmol, 1.2 eq) were added, and stirred for 3 h at room temperature. Compound 4 (30 mg, 46 μmol, 1.0 eq) and DIEA (15 μL, 93 μmol, 2.0 eq) were added and stirring was continued for 4 hours. The reaction solution was purified by HPLC to obtain a white solid (7.2 mg, 26%). MS (ESI): m/z found [M+1]+=591.4.
1H NMR (400 MHz, DMSO-d6): δ 8.69-8.44 (m, 1H), 7.79 (d, J=10.9 Hz, 1H), 7.30 (s, 1H), 6.52 (s, 1H), 5.52 (dt, J=9.1, 4.7 Hz, 1H), 5.41 (d, J=4.2 Hz, 2H), 5.20 (d, J=2.5 Hz, 2H), 4.40 (t, J=5.8 Hz, 1H), 4.25 (dd, J=8.1, 4.0 Hz, 1H), 4.07 (dd, J=20.7, 5.3 Hz, 1H), 4.01 (d, J=5.1 Hz, 1H), 3.50-3.44 (m, 2H), 3.15 (d, J=4.0 Hz, 2H), 2.38 (s, 3H), 2.16-1.96 (m, 4H), 1.93-1.76 (m, 4H), 0.84 (t, J=7.4 Hz, 3H) ppm.
EXD (25 mg, 47 gmol, 1.0 eq) and compound 6a (8.1 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL), and HATU (20 mg, 52 μmol, 1.1 eq) and DIEA (16 μL, 0.1 mmol, 2.1 eq) were added, and stirred for 2 h at room temperature. The reaction solution was purified by reversed-phase column chromatography (55% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (10.4 mg, 38%), MS (ESI): m/z found [M+1]+=590.0.
1H NMR (400 MHz, DMSO-d6): δ 8.86 (d, J=8.4 Hz, 1H), 7.95 (d, J=2.2 Hz, 1H), 7.88-7.74 (m, 2H), 7.30 (s, 1H), 7.01 (d, J=8.5 Hz, 1H), 6.52 (s, 1H), 5.76 (q, J=6.4 Hz, 1H), 5.38 (s, 2H), 5.25-5.05 (m, 2H), 3.26-3.12 (m, 2H), 3.26-3.12 (m, 2H) 5.76 (q, J=6.4 Hz, 1H), 5.38 (s, 2H), 5.25-5.05 (m, 2H), 3.26-3.12 (m, 2H), 2.42 (s, 3H), 2.31-2.14 (m, H), 1.85 (hept, J=7.1 Hz, 2H), 0.86 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 7a (8.1 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (16 μL, 118 μmol, 2.1 eq) were added, and stirred for 2 h at room temperature. The reaction solution was purified by reversed-phase column chromatography (55% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (5.2 mg, 19%), MS (ESI): m/z found [M+1]+=590.0.
1H NMR (400 MHz, DMSO-d6): δ 10.59 (s, 1H), 9.01 (d, J=8.4 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.54 (d, J=2.0 Hz, 1H), 7.41 (d, J=8.3 Hz, 1H), 7.34 (dd, J=8.3, 1.9 Hz, 1H), 7.30 (s, 1H), 6.54 (s, 1H), 5.75 (q, J=6.9, 6.2 Hz, 1H), 5.38 (s, 2H), 5.22 (d, J=18.8 Hz, 11H), 5.08 (d, J=18.9 Hz, 11H), 3.26-3.12 (m, 2H), 2.41 (s, 3H 2.41 (s, 3H), 2.31-2.20 (m, 2H), 1.90-1.78 (m, J=7.2 Hz, 2H), 0.86 (t, J=7.3 Hz, 3H).
p-Nitrophenyl chloroformate 8b (141 mg, 0.7 mmol, 1.0 eq) was dissolved in dichloromethane (2.0 mL), and pyridine (56 μL, 0.7 mmol, 1.0 eq) and trifluoroethylamine (50 μL, 0.7 mmol, 1.0 eq) were added in an ice bath and stirred for 2 h at room temperature to obtain the reaction intermediate, MS (ESI): m/z found [M+1]+=265.0.
EXD (30 mg, 56 μmol, 1 eq) was dissolved in DMF (2.0 mL) and DIEA (19 μL, 0.11 mmol, 2.0 eq) was added. After the reaction solution was clarified, 0.2 mL of the reaction solution intermediate was added and stirred for 1 h at room temperature. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and then purified by column chromatography (8% MeOH in EA) to obtain a white solid (20.5 mg, 65%), MS (ESI): m/z found [M+1]+=561.2.
1H NMR (400 MHz, DMSO-d6): δ 8.67 (d, J=10.8 Hz, 1H), 7.30 (s, 1H), 7.12 (d, J=8.8 Hz, 1H), 6.68 (t, J=6.0 Hz, 1H), 6.52 (br, 1H), 5.42 (s, 2H), 5.40-5.35 (m, 1H), 5.27 (s, 2H), 3.94-3.85 (m, 2H), 3.17 (t, J=6.0 Hz, 2H), 2.40 (s, 3H), 2.18 (t, J=6.0 Hz, 2H), 1.92-1.83 (m, 2H), 0.87 (t, J=6.0 Hz, 2H), 1.92-1.83 (m, 2H), 1.92-1.83 (m, 2H), 1.92-1.83 (m, 2H) 0.87 (t, J=4.0 Hz, 3H).
Compound 9a (100 mg, 0.45 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (40 μL, 0.49 mmol, 1.1 eq) and compound 8b (100 mg, 0.49 mmol, 1.1 eq) were added in an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (25% EA in PE) to obtain a yellow solid (120 mg, 69%). MS (ESI): m/z found [M+1]+388.1.
EXD (30 mg, 56 μmol, 1.0 eq) was dissolved in DMF (1.0 mL) and DMAP (13.8 mg, 0.11 mmol, 2.0 eq) was added. After the reaction solution was clarified, the compound 9b was added (22 mg, 56 μmol, 1.0 eq) and stirred at 80° C. for 5 hours. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and then purified by column chromatography (5% MeOH in CH2C2) to give a white solid (26 mg, 67%), MS (ESI): m/z found [M−55]+=628.3.
Compound 9b (25 mg, 37 μmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added, and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (33% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (15 mg, 59%) with MS (ESI): m/z found [M+1]+=584.2.
1H NMR (400 MHz, DMSO-d6): δ 7.67 (d, J=10.9 Hz, 1H), 7.20 (s, 1H), 7.06 (d, J=7.9 Hz, 2H), 6.76 (d, J=7.9 Hz, 2H), 6.64 (d, J=8.9 Hz, 1H), 6.42 (s, 1H), 6.25 (t, J=5.9 Hz, 1H), 5.34 (s, 2H), 5.30-5.23 (m, 2H), 5.15 (d, J=19.2 Hz, 1H), 4.14-4.02 (m, 2H), 3.05 (q, J=6.2 Hz, 3H), 2.28 (s, 3H), 2.04 (tt, J=6.9 Hz, 1H) 2.04 (tt, J=12.0, 6.5 Hz, 2H), 1.84-1.69 (m, 2H), 0.77 (t, J=7.3 Hz, 3H).
Compound 10a (100 mg, 0.50 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (44 μL, 0.55 mmol, 1.1 eq) and compound 8b (111 mg, 0.55 mmol, 1.1 eq) were added in an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (33% EA in PE) to obtain a yellow solid (108 mg, 59%).
MS (ESI): m/z found [M−99]+=266.2.
EXD (20 mg, 38 μmol, 1 eq) was dissolved in DMF (0.8 mL), and DMAP (9.2 mg, 75 μmol, 2.0 eq) was added. After the reaction solution was clarified, compound 10a (14 mg, 38 μmol, 1.0 eq) was added and stirred at 60° C. for 3 hours. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and then purified by column chromatography (6% MeOH in CH2Cl2) to obtain a white solid (16 mg, 67%), MS (ESI): m/z found [M−55]+=606.3.
Compound 10c (15 mg, 23 μmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added, and stirred for 3 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (33% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (9.1 mg, 59%) with MS (ESI): m/z found [M+1]+=562.2.
1H NMR (400 MHz, DMSO-d6): δ 8.43 (s, 1H), 8.23 (s, 1H), 7.73 (d, J=10.9 Hz, 1H), 7.26 (s, 1H), 6.54 (d, J=8.9 Hz, 1H), 6.49 (s, 1H), 6.26 (d, J=7.6 Hz, 1H), 5.37 (s, 2H), 5.30 (d, J=19.0 Hz, 1H), 3.76-3.68 (m, 1H), 5.12 (d, J=19.2 Hz, 1H), 3.76-3.68 (m, 19.2 Hz, 1H). 2H), 5.30 (d, J=19.0 Hz, 1H), 5.12 (d, J=19.2 Hz, 1H), 3.76-3.68 (m, 1H), 3.26-3.09 (m, 4H), 3.02-2.91 (m, 2H), 2.33 (s, 3H), 2.18-2.15 (m, 3H), 2.18-2.15 (m, 3H), 2.33 (s, 3H), 2.33 (s, 3H), 2.33 (s, 3H) 2.18-2.15 (m, 1H), 2.07 (dd, J=9.1, 4.4 Hz, 1H), 2.01-1.90 (m, 2H), 1.81 (dq, J=14.1, 7.0 Hz, 2H), 1.59-1.46 (m 2H), 0.83 (t, J=7.3 Hz, 3H).
Compound 11a (100 mg, 0.5 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (44 μL, 0.55 mmol, 1.1 eq) and compound 8b (111 mg, 0.55 mmol, 1.1 eq) were added to the mixture in an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (33% EA in PE) to obtain a yellow solid (110 mg, 60%). MS (ESI): m/z found [M−99]+=266.2.
EXD (20 mg, 38 μmol, 1 eq) was dissolved in DMF (0.8 mL), and DMAP (9.2 mg, 75 μmol, 2.0 eq) was added. After the reaction solution was clarified, compound 11b (14 mg, 38 μmol, 1.0 eq) was added and stirred at 60° C. for 3 h. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and purified by column chromatography (6% MeOH in CH2Cl2) to obtain a white solid (20 mg, 80%), MS (ESI): m/z found [M−55]+=606.3.
Compound 11c (20 mg, 30 μmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (0.5 mL) was added and stirred for 5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (33% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (12 mg, 59%) with MS (ESI): m/z found [M+1]+=562.2.
1H NMR (400 MHz, DMSO-d6): δ 8.55 (s, 1H), 8.46 (d, J=9.8 Hz, 1H), 7.69 (d, J=10.9 Hz, 1H), 7.23 (s, 1H), 6.68 (d, J=8.9 Hz, 1H), 6.46 (s, 1H), 6.16 (d, J=7.5 Hz, 1H), 5.33 (s, 2H), 5.31-5.21 (m, 2H), 5.09 (d, J=19.0 Hz, 1H), 3.84-3.75 (m, 1H), 3.16-3.01 (m, 4H), 2.77-2.61 (m, 2H), 2.30 (s, 3H), 2.16-2.09 (m, 1H), 2.06-1.97 (m, 1H), 1.85-1.70 (m, 4H), 1.62-1.51 (m, 1H), 1.42-1.31 (m, 1H), 0.78 (t, J=7.3 Hz, 3H).
Compound 12a (100 mg, 0.5 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (44 μL, 0.55 mmol, 1.1 eq) and compound 8b (111 mg, 0.55 mmol, 1.1 eq) were added under an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (33% EA in PE) to obtain a yellow solid (115 mg, 63%). MS (ESI): m/z found [M−99]+=266.2.
EXD (20 mg, 38 μmol, 1 eq) was dissolved in DMF (0.8 mL), and DMAP (9.2 mg, 75 μmol, 2.0 eq) was added. After the reaction solution was clarified, compound 12b (14 mg, 38 μmol, 1.0 eq) was added and stirred at 60° C. for 3 h. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and purified by column chromatography (6% MeOH in CH2Cl2) to obtain a white solid (15 mg, 64%), MS (ESI): m/z found [M−55]+=606.3.
Compound 12c (15 mg, 23 μmol) was dissolved in tetrahydrofuran (0.5 mL), and added to dissolve in dichloromethane (2 mL). Trifluoroacetic acid (0.5 mL) was added and stirred at room temperature for 5 hours. Concentrated and then purified by reversed-phase column chromatography (33% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (10 mg, 65%), MS (ESI): m/z found [M+1]+=562.2.
1H NMR (400 MHz, DMSO-d6): δ 8.58 (s, 1H), 8.49 (s, 1H), 7.77 (d, J=10.9 Hz, 1H), 7.28 (s, 1H), 6.72 (d, J=8.8 Hz, 1H), 6.51 (s, 1H), 6.09 (d, J=7.4 Hz, 1H), 5.39 (s 2H), 5.36-5.20 (m, 2H), 5.20 (d, J=19.2 Hz, 1H), 3.89-3.74 (m, 1H), 3.25-3.10 (m, 4H), 2.80-2.62 (m, 2H), 2.37 (s, 3H), 2.20-2.06 (m, 2H), 1.90-1.78 (m, 4H), 1.67-1.56 (m, 1H), 1.44-1.36 (m, 1H), 0.84 (t, J, 1H), 1.44-1.36 (m, 1H), 1.44-1.36 (m, 1H), 0.84 (t, J, 1H) 0.84 (t, J=7.3 Hz, 3H).
Compound 13a (100 mg, 0.54 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (48 μL, 0.59 mmol, 1.1 eq) and compound 8b (119 mg, 0.59 mmol, 1.1 eq) were added in an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (30% EA in PE) to obtain a yellow solid (120 mg, 64%). MS (ESI): m/z found [M−55]+296.2.
EXD (30 mg, 56 μmol, 1.0 eq) was dissolved in DMF (1.0 mL) and DMAP (14 mg, 0.11 mmol, 2.0 eq) was added. After the reaction solution was clarified, compound 13b (20 mg, 56 μmol, 1.0 eq) was added and stirred at 65° C. for 6 hours. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and purified by column chromatography (10% MeOH in CH2Cl2) to obtain a yellow solid (25.6 mg, 70%), MS (ESI): m/z found [M−55]+=592.3.
Compound 13c (25 mg, 39 μmol) was dissolved in dichloromethane (2 mL). Trifluoroacetic acid (0.5 mL) was added, and stirred for 5 h at room temperature. Concentrated and purified by reversed-phase column chromatography (30% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (15 mg, 60%), MS (ESI): m/z found [M+1]+=548.2.
1H NMR (400 MHz, DMSO-d6): δ 8.90 (s, 1H), 8.81 (s, 1H), 7.69 (d, J=10.9 Hz, 1H), 7.23 (s, 1H), 6.91 (d, J=8.9 Hz, 1H), 6.55 (d, J=6.4 Hz, 1H), 6.46 (s, 1H), 5.34 (s 2H), 5.31-5.23 (m, 2H), 5.10 (d, J=19.1 Hz, 1H), 4.21 (m, 1H), 3.25-3.20 (m, 1H), 3.18-3.03 (m, 4H), 3.18-3.03 (m, 4H), 3.03-2.93 (m, 1H), 2.30 (s, 3H), 2.14-2.01 (m, 3H), 1.83-1.72 (m, 3H), 0.79 (t, J=7.3 Hz, 3H).
Compound 14a (100 mg, 0.54 mmol, 1.0 eq) was dissolved in dichloromethane (2 mL). Pyridine (48 μL, 0.59 mmol, 1.1 eq) and compound 8b (119 mg, 0.59 mmol, 1.1 eq) were added in an ice bath and stirred for 2 h at room temperature. Water was added, extracted with dichloromethane, dried and concentrated and then purified by column chromatography (30% EA in PE) to obtain a yellow solid (107 mg, 57%). MS (ESI): m/z found [M−55]+296.2.
EXD (30 mg, 56 μmol, 1.0 eq) was dissolved in DMF (1.0 mL) and DMAP (14 mg, 0.11 mmol, 2.0 eq) was added. After the reaction solution was clarified, compound 14b (20 mg, 56 μmol, 1.0 eq) was added and stirred at 65° C. for 6 hours. The reaction solution was quenched with water, extracted with ethyl acetate, dried and concentrated and then purified by column chromatography (6% MeOH in CH2C2) to obtain a yellow solid (25 mg, 69%), MS (ESI): m/z found [M−55]+=592.3.
Compound 14c (25 mg, 39 μmol) was dissolved in dichloromethane (2 mL). trifluoroacetic acid (0.5 mL) was added, and stirred for 3 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (30% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (14 mg, 56%), MS (ESI): m/z found [M+1]+=548.2.
1H NMR (400 MHz, DMSO-d6): δ 8.76 (s, 1H), 8.69 (s, 1H), 7.72 (d, J=10.9 Hz, 1H), 7.27 (s, 1H), 6.82 (d, J=9.0 Hz, 1H), 6.50 (s, 1H), 6.44 (d, J=6.2 Hz, 1H), 5.37 (s, 2H), 5.35-5.24 (m, 2H), 5.11 (d, J=19.0 Hz, 1H), 4.24 (h, J=6.2 Hz, 1H), 4.24 (h, J=6.2 Hz, 1H) 2H), 5.35-5.24 (m, 2H), 5.11 (d, J=19.0 Hz, 1H), 4.24 (h, J=6.2 Hz, 1H), 3.28-3.22 (m, 2H), 3.14 (ddt, J=19.7, 14.8, 7.2 Hz, 4H), 2.33 (s, 3H) 2.33 (s, 3H), 2.20-2.09 (m, 2H), 2.06 (tdt, J=8.1, 3.8 Hz, 1H), 1.89-1.73 (m, 3H), 0.82 (t, J=7.3 Hz, 3H).
Compound 15a (50 mg, 0.23 mmol, 1.0 eq) was dissolved in methanol (1 mL). (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (40% EA in PE) to obtain a white solid (65 mg, 88%). MS (ESI): m/z found [M−1]−=312.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 15b (15 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (60% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (29 mg, 85%). MS (ESI): m/z found [M−55]+=675.3.
Compound 15c (29 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (34% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (18 mg, 70%). MS (ESI): m/z found [M+1]+=631.2.
1H NMR (400 MHz, DMSO-d6): δ 9.01 (d, J=8.4 Hz, 1H), 7.96 (d, J=8.5 Hz, 2H), 7.82 (d, J=11.0 Hz, 1H), 7.66 (d, J=8.5 Hz, 2H), 7.50 (d, J=8.5 Hz, 2H), 7.31 (s, 1H), 7.25-6.96 (m, 2H), 6.77 (d, J=8.2 Hz, 2H), 6.55 (s, 1H), 5.81 (d, J=8.2 Hz, 1H) 7.25-6.96 (m, 2H), 6.77 (d, J=8.2 Hz, 2H), 6.55 (s, 1H), 5.81 (dd, J=13.2, 6.4 Hz, 1H), 5.37 (s, 2H), 5.24 (d, J=18.8 Hz, 1H), 5.13 (d, J=18.9 Hz 1H), 2.42 (s, 3H), 2.34-2.23 (m, 2H), 1.83 (td, J=14.4, 7.0 Hz, 2H), 1.52 (d, J=7.0 Hz, 1H), 0.86 (t, J=7.3 Hz, 3H).
Compound 16a (50 mg, 0.27 mmol, 1.0 eq) was dissolved in methanol (1 mL), and (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (50% EA in PE) to obtain a white solid (70 mg, 91%). MS (ESI): m/z found [M−1]−=284.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 16b (13.5 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (65% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (28 mg, 85%). MS (ESI): m/z found [M−55]+=647.2.
Compound 16c (28 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (35% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (18.6 mg, 65%). MS (ESI): m/z found [M+1]+=603.2.
1H NMR (400 MHz, DMSO-d6): δ 8.61 (d, J=8.6 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 7.09 (d, J=8.2 Hz, 1H), 6.70 (d, J=2.0 Hz, 1H), 6.57 (dd, J=8.2, 2.0 Hz, 1H), 5.59-5.52 (m, 1H), 5.44 (s, 2H), 5.28 (d, J=18.9 Hz, 1H), 5.20 (d, 2H), 5.20 (d, J=18.9 Hz, 1H) 1H), 5.59-5.52 (m, 1H), 5.44 (s, 2H), 5.28 (d, J=18.9 Hz, 1H), 5.20 (d, J=19.0 Hz, 1H), 3.49 (s, 2H), 3.22-3.15 (m, 2H), 2.41 (s 3H), 2.22-2.08 (m, 2H), 1.94-1.82 (m, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 17a (50 mg, 0.27 mmol, 1.0 eq) was dissolved in methanol (1 mL), and (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (40% EA in PE) to obtain a white solid (68 mg, 88%). MS (ESI): m/z found [M−1]−=284.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 17b (13.5 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL), and HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (65% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (23 mg, 70%). MS (ESI): m/z found [M−55]+=647.2.
Compound 17c (23 mg, 33 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (35% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (15.5 mg, 66%). MS (ESI): m/z found [M+1]+=603.2.
1H NMR (400 MHz, DMSO-d6): δ 8.58 (d, J=8.6 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (s, 1H), 7.05 (d, J=1.8 Hz, 1H), 6.93 (dd, J=8.2, 1.9 Hz, 1H), 6.71 (d, J=8.2 Hz, 1H), 6.53 (s, 1H), 5.55-5.49 (m, 1H), 5.44 (s, 2H), 5.25 (d, J=18.9 Hz 1H), 6.53 (s, 1H), 5.55-5.49 (m, 1H), 5.44 (s, 2H), 5.25 (d, J=18.9 Hz, 1H), 5.05 (d, J=19.0 Hz, 1H), 3.31 (s, 2H), 3.17 (s, 2H), 2.41 (s, 3H). 2.13 (ddd, J=16.6, 9.5, 5.7 Hz, 2H), 1.93-1.81 (m, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 18a (50 mg, 0.28 mmol, 1.0 eq) was dissolved in methanol (1 mL), and (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (25% EA in PE) to obtain a white solid (65 mg, 84%). MS (ESI): m/z found [M−1]−=280.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 18b (13.2 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (64% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (21 mg, 65%). MS (ESI): m/z found [M−55]+=643.4.
Compound 18c (21 mg, 30 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (37% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (13.2 mg, 62%). MS (ESI): m/z found [M+1]+=598.3.
Compound 19a (50 mg, 0.28 mmol, 1.0 eq) was dissolved in methanol (1 mL). (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (25% EA in PE) to obtain a white solid (70 mg, 900). MS (ESI): m/z found [M−1]−=280.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 19b (13.2 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (64% CH3CN in H2O, 0.0500 HCOOH) to obtain a white solid (20 mg, 61%). MS (ESI): m/z found [M−55]+=643.4.
Compound 19c (20 mg, 29 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (37% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (10.8 mg, 53%). MS (ESI): m/z found [M+1]+=598.3.
1H NMR (400 MHz, DMSO-d6): δ 8.50 (d, J=8.6 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 7.06 (d, J=8.0 Hz, 1H), 6.53 (s, 1H), 6.48 (d, J=7.8 Hz, 1H), 5.59-5.49 (m, 1H), 5.44 (s, 2H), 5.22 (dd, J=34.2, 19.0 Hz, 2H), 3.65 (s, 3H), 3.38 (s, 2H), 3.22-3.15 (m, 2H), 2.41 (s, 3H), 2.22-2.08 (m, 2H), 1.88 (ddq, J=28.9, 14.3, 7.2 Hz, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 20a (50 mg, 0.3 mmol, 1.0 eq) was dissolved in methanol (1 mL) and tetrahydrofuran (1 mL). (Boc)2O (82 μL, 0.35 mmol, 1.2 eq) and DIEA (60 μL, 0.35 mmol, 1.2 eq) were added, and stirred for 12 h at room temperature. The reaction solution was diluted with water, extracted with ethyl acetate, dried and concentrated and then purified by column chromatography (20% EA in PE) to obtain a white solid (70 mg, 880). MS (ESI): m/z found [M−1]−==268.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 20b (12.7 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature.
The reaction solution was directly purified by reversed-phase column chromatography (60% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (20 mg, 670%). MS (ESI): m/z found [M−55]+=631.3.
Compound 20c (20 mg, 29 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (37% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (14.5 mg, 71%). MS (ESI): m/z found [M+1]+=587.3.
Compound 21a (50 mg, 0.3 mmol, 1.0 eq) was dissolved in methanol (1 mL) and tetrahydrofuran (1 mL). (Boc)2O (82 μL, 0.35 mmol, 1.2 eq) and DIEA (60 μL, 0.35 mmol, 1.2 eq) were added, and stirred for 12 h at room temperature. The reaction solution was diluted with water, extracted with ethyl acetate, dried and concentrated, and then purified by column chromatography (20% EA in PE) to obtain a white solid (75 mg, 94%). MS (ESI): m/z found [M−1]−=268.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 21b (12.7 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (60% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (22 mg, 74%). MS (ESI): m/z found [M−55]+=631.3.
Compound 21c (22 mg, 29 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (37% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (15.5 mg, 69%). MS (ESI): m/z found [M+1]+=587.3.
1H NMR (400 MHz, DMSO-d6): δ 8.62 (d, J=8.5 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (s, 1H), 7.23-6.98 (m, 1H), 7.03 (t, J=8.6 Hz, 1H), 6.42 (t, J=9.2 Hz, 2H), 5.59-5.50 (m, 1H), 5.44 (s, 2H), 5.27 (d, J=18.9 Hz, 1H), 5.15 (d, J=19.0 Hz, 1H), 3.37 (d, J=5.0 Hz, 2H), 3.23-3.14 (m 2H), 2.41 (s, 3H), 2.22-2.05 (m, 2H), 1.87 (tt, J=14.0, 7.0 Hz, 2H), 0.88 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 22a (7.2 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (34% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (20 mg, 75%). MS (ESI): m/z found [M+1]+=570.0.
1HNMR (400 MHz, DMSO-d6) δ 9.22 (s, 1H), 8.59 (d, J=8.6 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (s, 1H), 7.07 (d, J=8.5 Hz, 2H), 6.72-6.64 (m, 2H), 6.53 (s, 1H), 5.52 (dt, J=9.0, 4.8 Hz, 1H), 5.44 (s, 2H), 5.25 (d, J=18.9 Hz, 1H), 5.11 (d, J=18.9 Hz, 1H), 3.17 (t, J=6.3 Hz, 2H), 2.41 (d, J=1.8 Hz, 2H), 2.16-2.09 (m, 2H), 1.95-1.80 (m, 3H), 0.88 (t, J=7.3 Hz, 3H).
Compound 23a (50 mg, 0.33 mmol, 1.0 eq) was dissolved in methanol (1 mL). (Boc)2O (0.5 mL) was added and stirred for 6 h at room temperature.
Then then reaction solution was purified by column chromatography (25% EA in PE) to obtain a white solid (75 mg, 90%). MS (ESI): m/z found [M−1]−=250.2.
EXD (25 mg, 47 μmol, 1 eq) and compound 23b (11.8 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (63% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (20 mg, 64%). MS (ESI): m/z found [M−55]+=613.2.
Compound 23c (20 mg, 30 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (35% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (14.3 mg, 70%). MS (ESI): m/z found [M+1]+=569.2.
1H NMR (400 MHz, DMSO-d6): δ 8.72 (d, J=8.5 Hz, 1H), 7.82 (d, J=10.9 Hz, 1H), 7.33 (d, J=1.9 Hz, 1H), 7.18 (t, J=7.7 Hz, 1H), 6.93 (s, 1H), 6.85 (dd, J=20.0, 6.9 Hz 2H), 6.56 (s, 1H), 5.58-5.50 (m, 1H), 5.45 (d, J=4.1 Hz, 2H), 5.21 (dd, J=16.4, 6.1 Hz, 2H), 3.46 (s, 2H), 3.18 (s, 2H), 2.41 (s, 3H), 2.14 (dd, J=16.0, 11.2 Hz, 2H), 1.93-1.81 (m, 2H), 0.88 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1 eq) and compound 24a (11.8 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (63% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (22 mg, 70%). MS (ESI): m/z found [M−55]+=613.2.
Compound 24b (22 mg, 33 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentration was followed by reversed-phase column chromatography purification (35% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (14.4 mg, 64%). MS (ESI): m/z found [M+1]+=569.2.
1H NMR (400 MHz, DMSO-d6): δ 8.78 (d, J=8.5 Hz, 1H), 7.83 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 7.10 (t, J=7.7 Hz, 2H), 6.90 (d, J=7.3 Hz, 1H), 6.81 (s, 1H), 6.55 (s, 1H), 5.54 (dt, J=8.5, 4.2 Hz, 1H), 5.43 (s, 2H), 5.30 (d, J=18.9 Hz, 1H), 5.15 (d, J=18.9 Hz, 1H), 3.48 (s, 2H), 3.18 (s, 2H), 2.41 (s, 3H), 2.22-2.02 (m, 2H), 1.87 (tt, J=14.0, 7.2 Hz, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 25a (50 mg, 0.3 mmol, 1.0 eq) was dissolved in tetrahydrofuran (1 mL). (Boc)2O (83 μL, 0.36 mmol, 1.2 eq) was added, and stirred for 16 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (20% EA in PE) to obtain a white solid (60 mg, 75%). MS (ESI): m/z found [M−1]−=264.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 25b (12.5 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (57% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (20 mg, 63%). MS (ESI): m/z found [M−55]+=627.3.
Compound 25c (20 mg, 29 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (25% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (12 mg, 59%). MS (ESI): m/z found [M+1]+=583.3.
1H NMR (400 MHz, DMSO-d6): δ 8.45 (d, J=8.7 Hz, 1H), 7.80 (d, J=10.9 Hz, 11H), 7.31 (s, 11H), 7.17 (d, J=8.0 Hz, 2H), 6.96 (d, J=7.8 Hz, 2H), 6.54 (s, 1H), 5.54 (dt, J=9.1, 4.8 Hz, 1H), 5.44 (s, 2H), 5.23 (d, J=18.9 Hz, 1H), 5.10 (d, J=19.0 Hz, 1H), 3.16-3.07 (m, 2H), 2.97-2.89 (m, 2H), 2.89-2.82 (m, 2H), 2.40 (d, J=1.9 Hz, 3H), 2.07 (dt, J=13.8, 8.1 Hz, 2H), 1.92-1.81 (m, 2H), 1.16 (t, J=7.3 Hz, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 26a (50 mg, 0.28 mmol, 1.0 eq) was dissolved in tetrahydrofuran (1 mL). (Boc)2O (77 μL, 0.33 mmol, 1.2 eq) was added, and stirred for 18 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (25% EA in PE) to obtain a white solid (65 mg, 83%). MS (ESI): m/z found [M−1]−=278.1.
EXD (25 mg, 47 μmol, 1 eq) and compound 26b (13.1 mg, 47 μmol, 1 eq) were dissolved in DMF (1 mL). HATU (26.8 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 118 μmol, 2.5 eq) were added, and stirred for 2 h at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (57% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (18 mg, 55%). MS (ESI): m/z found [M−55]+=641.3.
Compound 26c (18 mg, 26 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (25% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (12 mg, 65%). MS (ESI): m/z found [M+1]+=597.3.
1H NMR (400 MHz, DMSO-d6): δ 8.48 (t, J=7.6 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (d, J=2.3 Hz, 1H), 7.28 (dd, J=8.3, 3.9 Hz, 2H), 7.16 (d, J=8.3 Hz, 2H), 5.58 (dt J=9.5, 5.1 Hz, 1H), 5.42 (s, 2H), 5.25 (d, J=19.0 Hz, 1H), 5.16 (d, J=18.9 Hz, 1H), 3.17 (s, 2H), 2.60 (t, J=7.7 Hz, 2H), 2.40 (d, J=1.8 Hz, 3H), 2.19 (t, J=7.5 Hz, 2H), 2.13 (dd, J=11.9, 5.9 Hz, 2H), 1.95-1.78 (m, 4H), 1.54-1.38 (m, 1H), 1.28-1.23 (m, 1H), 1.07 (dt, J=17.7, 5.7 Hz, 1H), 0.87 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 27a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was purified by reverse column chromatography (65% CH3CN in H2O, 0.05% HCOOH). The reaction solution was directly purified by reversed-phase column chromatography (65% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (24 mg, 79%). MS (ESI): m/z found [M+1]+=647.2.
Compound 27b (24 mg, 37 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentration was followed by reversed-phase column chromatography purification (35% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (16 mg, 65%). MS (ESI): m/z found [M+1]+=547.2.
1H NMR (400 MHz, DMSO-d6): δ 8.71 (d, J=8.6 Hz, 1H), 8.57 (s, 2H), 7.82 (d, J=10.9 Hz, 1H), 7.32 (s, 1H), 6.56 (s, 1H), 5.56 (dt, J=9.0, 4.7 Hz, 1H), 5.43 (s, 2H), 5.26, 5.16 (ABq, 2H, J=18.8 Hz), 3.35-3.28 (m, 1H), 3.22-3.07 (m, 4H), 2.94 (s, 1H), 2.65 (dq, J=10.0, 5.9, 5.0 Hz, 1H), 2.41 (s 3H), 2.14 (tt, J=8.4, 5.8, 5.4 Hz, 2H), 1.98-1.78 (m, 4H), 1.64 (td, J=17.9, 15.7, 8.4 Hz, 2H), 0.87 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 28a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (62% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (26 mg, 86%). MS (ESI): m/z found [M−55]+=591.2.
Compound 28b (26 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (33% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (18 mg, 73%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 8.75 (d, J=8.7 Hz, 1H), 8.63-8.46 (m, 2H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (s, 1H), 6.54 (s, 1H), 5.57 (dt, J=9.2, 4.9 Hz. 1H), 5.42 (s, 2H), 5.23 (d, J=18.8 Hz, 1H), 5.06 (d, J=18.8 Hz, 1H), 3.29 (d, J=12.3 Hz, 1H), 3.20-3.04 (m, 4H), 2.92 (q, J=10.7 Hz, 1H), 2.65 (tt, J=10.1, 4.1 Hz, 1H), 2.40 (d, J=1.9 Hz, 3H), 2.15 (t, J=6.0 Hz, 2H), 1.97-1.74 (m, 4H), 1.69-1.50 (m, 2H), 0.88 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 29a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (62% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (26 mg, 86%). MS (ESI): m/z found [M−55]+=591.2.
Compound 29b (26 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (31% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (22 mg, 83%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 9.06 (d, J=8.7 Hz, 1H), 9.00 (d, J 10.9 Hz, 1H), 8.88 (q, J=10.7 Hz, 1H), 7.84 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 6.56 (s, 1H), 5.65 (dt J=8.8, 4.4 Hz, 1H), 5.43 (s, 2H), 5.29 (s, 1H), 5.05 (s, 1H), 3.78-3.71 (m, 1H), 3.30-3.20 (m, 2H), 3.19-3.09 (m 1H), 2.99-2.86 (m, 1H), 2.42 (s, 3H), 2.17 (tq, J=13.8, 8.8, 6.6 Hz, 2H), 2.04 (d, J=13.0 Hz, 1H), 1.96-1.77 (m, 2H), 1.76-1.65 (m, 2H), 1.57 (qd, J=13.1, 3.6 Hz, 2H), 1.38 (d, J=12.3 Hz, 1H), 0.88 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (62% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (28 mg, 92%). MS(ESI): m/z found [M+1]+=647.2.
Compound 30a (28 mg, 43 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (31% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (24 mg, 85%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 8.98-8.93 (m, 2H), 8.77 (q, J=10.9 Hz, 1H), 7.85 (d, J=10.9 Hz, 1H), 7.33 (s, 1H), 6.57 (s, 1H), 5.65 (dt, J=8.0, 3.5 Hz, 1H), 5.44 (s, 2H), 5.41 (d, J=19.0 Hz, 1H), 5.23 (d, J=19.0 Hz, 1H), 3.68-3.61 (m, 1H), 3.31-3.18 (m, 2H), 3.16-3.04 (m, 1H), 2.84 (q, J=11.5 Hz, 1H), 2.42 (s, 3H), 2.26-2.18 (m, 1H), 2.11 (td, J=13.2, 6.5 Hz, 2H), 1.92-1.76 (m, 3H), 1.73-1.52 (m, 3H), 1.43-1.36 (m, 1H), 0.86 (t, J=7.4 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (58% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (25 mg, 82%). MS (ESI): m/z found [M−99]+=547.2.
Compound 31b (25 mg, 39 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (30% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (20 mg, 66%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 8.62-8.55 (m, 2H), 8.31 (d, J=10.5 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.31 (s, 1H), 6.55 (s, 1H), 5.57 (dt, J=9.3, 5.1 Hz, 1H), 5.42 (s, 2H), 5.22, 5.09 (ABq, J=18.8 Hz, 2H), 3.38-3.28 (m, 2H), 3.17 (t, J=6.3 Hz, 2H), 2.94-2.79 (m, 2H), 2.49-2.44 (m, 1H), 2.40 (s, 3H), 2.14 (q, J=6.0 Hz, 2H), 1.98-1.76 (m, 6H), 0.87 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (58% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (26 mg, 86%). MS (ESI): m/z found [M−99]+=547.2.
Compound 32b (26 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (30% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (19 mg, 71%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 9.09-8.98 (m, 1H), 8.72 (d, J=8.6 Hz, 1H), 8.55-8.47 (m, 1H), 7.83 (d, J=10.9 Hz, 1H), 7.33 (s, 1H), 6.56 (s, 1H), 5.60 (dt, J=8.6, 4.3 Hz, 1H), 5.44 (s, 2H), 5.33 (d, J=8.6, 4.3 Hz, 1H), 5.44 (s, 2H), 5.44 (s, 2H) 6.56 (s, 1H), 5.60 (dt, J=8.6, 4.3 Hz, 1H), 5.44 (s, 2H), 5.33 (d, J=18.9 Hz, 1H), 5.23 (d, J=18.9 Hz, 1H), 3.79-3.71 (m, 1H), 3.21-3.13 (m, 4H), 2.62 (qd, J=16.1, 6.9 Hz, 2H), 2.42 (s, 3H), 2.25-2.06 (m, 3H), 1.96-1.77 (m, 4H), 1.54 (dq, J=13.0, 8.6 Hz, 1H), 0.87 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (56% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (24 mg, 79%). MS (ESI): m/z found [M−99]+=547.2.
Compound 33b (24 mg, 37 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (28% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (19 mg, 78%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 9.05 (s, 1H), 8.73 (d, J=8.7 Hz, 1H), 8.55 (s, 1H), 7.82 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 6.56 (s, 1H), 5.61 (dt, J=8.9, 4.6 Hz, 1H), 5.43 (s, 2H), 5.35-5.18 (m, 2H), 3.82-3.74 (m, 1H), 3.21-3.14 (m, 4H), 2.64 (d, J=6.9 Hz, 2H), 2.41 (s, 3H), 2.21-2.05 (m, 3H), 1.95-1.77 (m, 4H), 1.61-1.49 (m, 1H), 0.87 (t, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (57% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (26 mg, 86%). MS (ESI): m/z found [M−99]+=547.2.
Compound 34b (24 mg, 40 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (28% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (20 mg, 82%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 8.72 (s, 2H), 8.57 (d, J=8.7 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.32 (s, 1H), 6.56 (s, 1H), 5.58 (dt, J=9.3, 5.1 Hz, 1H), 5.43 (s, 2H), 5.27-5.12 (m, 2H), 3.43-3.36 (m, 1H), 3.27-3.09 (m, 4H), 2.87-2.78 (m, 4H), 3.27-3.09 (m, 4H), 2.87-2.78 (m, 4H) 5.27-5.12 (m, 2H), 3.43-3.36 (m, 1H), 3.27-3.09 (m, 4H), 2.87-2.78 (m, 1H), 2.64-2.55 (m, 1H), 2.43-2.29 (m, 5H), 2.16-2.08 (m, 3H), 1.87 (p, J=7.0 Hz, 2H), 1.61-1.51 (m, 1H), 0.87 (t, J=7.3 Hz 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (11 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL), and HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. The reaction solution was directly purified by reversed-phase column chromatography (57% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (22 mg, 72%). MS (ESI): m/z found [M+1]+=647.2.
Compound 35b (22 mg, 34 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (28% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (17 mg, 76%). MS (ESI): m/z found [M+1]+=547.4.
1H NMR (400 MHz, DMSO-d6): δ 8.75 (s, 1H), 8.67 (s, 1H), 8.58 (d, J=8.7 Hz, 11H), 7.81 (d, J=10.9 Hz, 11H), 7.31 (s, 11H), 6.55 (s, 11H), 5.58 (dt, J=9.3, 5.1 Hz, 1H), 5.43 (s, 2H), 5.29-5.10 (m, 2H), 3.40 (dtd, J=11.0, 6.8, 3.3 Hz, 1H) 5.43 (s, 2H), 5.29-5.10 (m, 2H), 3.40 (dtd, J=11.0, 6.8, 3.3 Hz, 1H), 3.27-3.07 (m, 4H), 2.85 (ddd, J=15.7, 12.0, 6.6 Hz, 1H), 2.60 (p, J=7.8 Hz, 1H), 2.40 (s, 3H), 2.36 (d, J=7.2 Hz, 2H), 2.18-2.04 (m, 3H), 1.86 (hept, J=7.2 Hz, 2H), 1.56 (dq, J=13.0, 8.8 Hz, 1H), 0.87 (d, J=7.3 Hz, 3H).
EXD (25 mg, 47 μmol, 1.0 eq) and compound 30a (10 mg, 47 μmol, 1.0 eq) were dissolved in DMF (1 mL). HATU (21 mg, 56 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.2 eq) were added, and stirred at 40° C. for 1.5 h. Water was added, extracted with ethyl acetate, washed with saturated saline, dried and concentrated and then purified by column chromatography (4% MeOH in CH2C2) to obtain a white solid (29 mg, 98%). MS (ESI): m/z found [M+1]+=633.4.
Compound 36b (29 mg, 46 μmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) and stirred for 2.5 h at room temperature. Concentrated and then purified by reversed-phase column chromatography (31% CH3CN in H2O, 0.1% TFA) to obtain a yellow solid (22 mg, 74%). MS (ESI): m/z found [M+1]+=533.4.
1H NMR (400 MHz, DMSO-d6): δ 9.18 (d, J=8.0 Hz, 1H), 9.00 (d, J=8.4 Hz, 1H), 8.72 (d, J=5.2 Hz, 1H), 7.85 (d, J=11.0 Hz, 1H), 7.33 (s, 1H), 6.57 (s, 1H), 5.62 (dt, J=8.1, 3.7 Hz, 1H), 5.44 (s, 2H), 5.39 (d, J=19.0 Hz, 1H), 5.27 (d, J=19.0 Hz, 1H), 4.14-4.07 (m, 1H), 3.33-3.19 (m, 3H), 3.17-3.06 (m, 1H), 2.43 (s, 3H), 2.29-2.20 (m, 2H), 2.11 (ddt, J=12.4, 8.1, 4.3 Hz, 1H), 1.96-1.79 (m, 5H), 0.86 (t, J=7.3 Hz, 3H).
To a solution of compound 37a (30 g, 96.5 mmol) and compound 37b (17.5 g, 96.5 mmol, 1.0 eq) in DMF (400 mL) were added HATU (36.7 g, 96.5 mmol, 1.0 eq) and DIEA (32 mL, 193 mmol, 2.0 eq). After stirring the reaction for 12 h at room temperature, the reaction solution was poured into water (1500 mL), extracted with ethyl acetate (500 mL×3), washed with saturated saline (500 mL), dried with anhydrous sodium sulfate, concentrated and then purified by column chromatography (25% EA in PE) to obtain a white solid (30 g, 70.9%). MS (ESI): m/z found [M−55]+=383.2.
A solution of TFA (150 mL) and H2O (10 mL) of compound 37c (30 g, 68.4 mmol) was reacted with stirring at room temperature for 4 h. The reaction solution was then concentrated and pulped with petroleum ether (500 mL) and ethyl acetate (50 mL) to obtain a white solid (34 g, 100%). MS (ESI): m/z found [M+1]+=383.2.
To a solution of compound 37e (30 g, 135 mmol) and HOSu (16.2 g, 141 mmol, 1.05 eq) in acetonitrile (300 mL) was added DCC (29 g, 141 mmol, 1.05 eq) in batches under an ice bath, and the reaction was stirred at room temperature for 2 h after the ice bath was withdrawn. At the end of the reaction the insoluble material was filtered out and used directly in the next step without concentration.
To a solution of compound 37f (solution above) and compound 37g (17.6 g, 141 mmol, 1.05 eq) in acetonitrile (400 mL) was added DIEA (44 mL, 282 mmol, 2.1 eq). After stirring the reaction for 2 h at room temperature, the reaction solution was concentrated, redissolved in CH2Cl2 (500 mL), washed with sodium bicarbonate solution (100 mL) and saturated saline (100 mL), dried with anhydrous sodium sulfate, concentrated and purified by normal-phase flash column chromatography (50% EA in PE) to obtain a white solid (27 g, 59.5%). MS (ESI): m/z found [M−55]+=281.0.
To a solution of MeOH (300 mL) and H2O (10 mL) of compound 37h (18 g, 53.6 mmol) was added 10% Pd/C (1.0 g, 55% H2O), evacuated and reacted under H2 atmosphere for 4 h. The insoluble material was filtered off and concentrated to obtain the colorless oily compound (10.8 g, 100%). MS (ESI): m/z found [2M+1]+=405.
To a solution of compound 37d (37.5 g, 98 mmol) and compound 37i (19.8 g, 98 mmol, 1.0 eq) in DMF (200 mL) were added HATU (37.3 g, 98 mmol, 1.0 eq) and DIEA (16 mL, 98 mmol, 1.0 eq). After stirring the reaction at room temperature for 4 h, the reaction solution was poured into water (1 L) to precipitate the white solid, filtered, washed with water, and pulped by methyl tert-butyl ether to obtain the white solid (40.0 g, 72%). MS (ESI): m/z found [M−55]+=511.2.
A solution of TFA (100 mL) and dichloromethane (300 mL) of compound 37i (40.0 g, 71 mmol) was reacted with stirring at room temperature for 3 h.
It was concentrated to obtain the yellow oil, and recrystallized with ethyl acetate (500 mL) to obtain a white solid (34.9 g, 97%). MS (ESI): m/z found [M−1]−=509.2.
Pb(OAc)4 (31.3 g, 72 mmol, 1.15 eq) was added to a solution of compound 37i (32.1 g, 63 mmol) and compound Cu(OAc)2 (4.33 g, 24 mmol, 0.38 eq) in DMF (300 mL) under nitrogen protection. The reaction solution was warmed up to 60° C. and after stirring the reaction at this temperature for 1 h, the reaction solution was poured into water (1.5 L) and a white solid was precipitated, which was filtered and dried to obtain a white solid (33.2 g, 94%). MS (ESI): m/z found [M+1]+=525.2.
To a solution of CH2Cl2 (500 mL) of compound 371 (32 g, 61 mmol) and compound 37m (17.3 mL, 122 mmol, 2.0 eq) was added TFA (120 mL). After stirring the reaction at room temperature for 1 h, the reaction was concentrated, dissolved in DMF, and purified by reversed-phase flash column chromatography (52% ACN in H2O, 0.05% HCOOH) to obtain compound 37n (16.0 g, 40%) as a white solid. MS (ESI): m/z found [M+23]+=653.2.
To a solution of compound 371 (15.2 g, 24 mmol) in DMF (80 mL) was added DBU (3.6 mL, 24 mmol, 1.0 eq). After stirring the reaction at room temperature for 1.5 h, the reaction solution was purified by reversed-phase flash column chromatography (26% ACN in H2O, 0.05% HCOOH) to obtain a white solid (7.8 g, 76%). MS (ESI): m/z found [M+1]+=409.2.
To a solution of compound 37o (4.0 g, 9.8 mmol) in MeOH (60 mL) was added 10% Pd/C (0.5 g), vacuumized, and reacted under a hydrogen atmosphere for 4 h. The insoluble material was filtered off and concentrated to obtain white (2.8 g, 90%). MS (ESI): m/z found [M+1]+=319.0.
To a solution of compound 37q (650 mg, 1.33 mmol) in ACN (10 mL) was added HOSu (161 mg, 1.40 mmol, 1.05 eq) and DCC (288 mg, 1.40 mmol, 1.05 eq) under an ice bath and the reaction was stirred at room temperature for 4 h. At the end of the reaction, the insoluble material was filtered out and concentrated to obtain the yellow oil compound (770 mg, 100%). MS (ESI): m/z found [M+1]+=585.0.
To a solution of compound 37r (777 mg, 1.33 mmol) and compound 37μ (423 mg, 1.33 mmol, 1.0 eq) in DMF (8 mL) was added DIEA (219 μuL, 1.33 mmol, 1.0 eq). After stirring the reaction at room temperature for 2 h, the compound was purified by reversed-phase flash column chromatography (40% ACN in H2O, 0.05% HCOOH) to obtain the white solid compound (702 mg, 67.0%). MS (ESI): m/z found [M+1]+=788.2.
To a solution of compound 37s (702 mg, 0.99 mmol) in DMF (6 mL) was added DBU (149 μL, 0.99 mmol, 1.0 eq). After 2 h of stirring at room temperature, compound 37t (315 mg, 0.99 mmol, 1.0 eq) was added. After stirring the reaction at room temperature for 1 h, the reaction solution was purified by reversed-phase flash column chromatography (23% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound.
MC-PEG4-AAA-OCH2COOH (600 mg, 79.8%). MS (ESI): m/z found [M+1]+=759.2.
To a solution of compound 38a (500 mg, 0.87 mmol) in ACN (10 mL) was added HOSu (110 mg, 0.96 mmol, 1.1 eq) and DCC (198 mg, 0.96 mmol, 1.1 eq) in an ice bath, and the reaction was stirred at room temperature for 4 h. At the end of the reaction, the insoluble material was filtered out, and the reaction mixture was concentrated to obtain the yellow oily compound (assumed 100% yield). MS (ESI): m/z found [M+1]+=673.0.
To a solution of compound 38b (585 mg, 0.87 mmol) and compound 37μ (277 mg, 0.87 mmol, 1.0 eq) in DMF (8 mL) was added DIEA (144 μL, 0.87 mmol, 1.0 eq). After stirring the reaction at room temperature for 2 h, the reaction mixture was purified by reversed-phase fast column chromatography (38% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound (580 mg, 72%). MS (ESI): m/z found [M−1]−=874.2.
To a solution of compound 38c (550 mg, 0.63 mmol) in DMF (6 mL) was added DBU (95 μL, 0.63 mmol, 1.0 eq). After 2 h of stirring at room temperature, 37t (194 mg, 0.63 mmol, 1.0 eq) was added. After 1 h of stirring at room temperature, the reaction solution was purified by reversed-phase flash column chromatography (24% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound MC-PEG6-AAA-OCH2 COOH (200 mg, 37%). MS (ESI): m/z found [M−1]−=845.4.
To a solution of ACN (10 mL) of compound 39a (650 mg, 0.98 mmol) was added HOSu(124 mg, 1.1 mmol, 1.1 eq) and DCC (222 mg, 1.1 mmol, 1.1 eq) under an ice bath, the ice bath was withdrawn, and the reaction was stirred at room temperature for 4 h. At the end of the reaction, the insoluble material was filtered out, and the reaction was concentrated to obtain the colorless oily compound 39b (746 mg, 100%). MS (ESI): m/z found [M+1]+=761.3.
To a solution of compound 39b (945 mg, 0.98 mmol) and compound 37μ (312 mg, 0.98 mmol, 1.0 eq) in DMF (8 mL) was added DIEA (162 μL, 0.98 mmol, 1.0 eq). After the reaction was stirred at room temperature for 2 h, the reaction mixture was purified by reversed-phase flash column chromatography (40% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound (815 mg, 86%). MS (ESI): m/z found [M−1]−=962.5.
To a solution of compound 39c (815 mg, 0.85 mmol) in DMF (6 mL) was added DBU (129 μL, 0.85 mmol, 1.0 eq). After 2 h of stirring at room temperature, compound 6 (262 mg, 0.85 mmol, 1.0 eq) was added. After 1 h of stirring reaction at room temperature, the reaction solution was purified by reversed-phase flash column chromatography (23% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound 39 (MC-PEG8-AAA-OCH2COOH) (330 mg, 41%). MS (ESI): m/z found [M−1]−=933.5.
To a solution of CH2Cl2 (90 mL) of compound 371 (7.7 g, 14.7 mmol) and compound 40a (5.1 mL, 73 mmol, 5.0 eq) was added TFA (16 mL). The reaction was stirred at room temperature for 1 h. The reaction was concentrated, then dissolved in DMF and purified by reversed-phase flash column chromatography (40% ACN in H2O, 0.05% HCOOH) to obtain the white solid compound (5.5 g, 67%). MS (ESI): m/z found [M+23]+=653.2.
To a solution of compound 40c (1.2 g, 2.46 mmol) in ACN (10 mL) was added DBU (367 μL, 2.46 mmol, 1.0 eq). After 2 h of stirring at room temperature, compound 37t (909 mg, 2.95 mmol, 1.2 eq) was added. After stirring the reaction at room temperature for 1 h, the reaction solution was concentrated and purified by reversed-phase flash column chromatography (28% ACN in H2O, HCOOH) to obtain the colorless oily compound (460 mg, 41%). MS (ESI): m/z found [M−1]−=457.2.
To a solution of compound 40d (460 mg, 1.0 mmol) in ACN (6 mL) was added (138 mg, 1.2 mmol, 1.2 eq) and DCC (248 mg, 1.40 mmol, 1.2 eq) under an ice bath, the ice bath was withdrawn, and the reaction was stirred at room temperature for 12 h. At the end of the reaction, the insoluble material was filtered out, and the reaction was concentrated to obtain the compound 40e as a colorless oil (550 mg, 99%). MS (ESI): m/z found [M+1]+=556.2.
To a solution of compound 40e (1.2 g, 2.46 mmol) in DMF (4 mL) was added DBU (71 μL, 0.47 mmol, 1.0 eq). After 1 h of stirring at room temperature, compound 40b (260 mg, 0.47 mmol, 1.0 eq) was added. After stirring the reaction at room temperature for 1 h, the reaction solution was purified by reversed-phase flash column chromatography (24% ACN in H2O, 0.05% HCOOH) to obtain the colorless oily compound 40 (MC-PEG4-AAA-SCH2COOH) (100 mg, 27.5%). MS (ESI): m/z found [M−1]−=773.2.
Compound 41a (10 g, 37.6 mmol) was dissolved in acetonitrile (100 mL). HOSu (4.76 g, 41.3 mmol, 1.1 eq) and EDCI-HCl (8.28 g, 43.2 mmol, 1.15 eq) were added and stirred at room temperature for 5 h. Approximately 10% of the feedstock remained for LCMS monitoring. Filtered and the filtrate was stirred at 0° C. for 5 h. A large amount of white solid was precipitated. The filtrate was filtered and the solid was dried to obtain a white solid (9.06 g, 87%). MS (ESI): m/z found [M+1]+=364.2.
Compound 41b (9.06 g, 25 mmol) and L-phenylalanine (4.54 g, 27.4 mmol, 1.1 eq) were dissolved in acetonitrile (50 mL) and water (50 mL), and triethylamine (3.8 mL, 27.4 mmol, 1.1 eq) was added and stirred at room temperature for 2 h. The reaction solution had turbidity changed to clarification. The pH was adjusted to 2-3 with hydrochloric acid. Then the reaction solution was continued stirring at room temperature for 6 hours, and precipitated white solid. Filtered, washed with water and the solid was dried to obtain a white solid (10.69 g, 100%). MS (ESI): m/z found [M+1]+=414.2.
DBU (951 μL, 6.1 mmol, 0.5 eq) was added to a solution of compound 41f (5.8 g, 12.2 mmol) in ACN (150 mL). After stirring the reaction for 2 h at room temperature, HOBt (2.1 g, 15.1 mmol, 1.2 eq) and compound 41d (5.8 g, 12.8 mmol, 1.05 eq) were added, the reaction solution was cooled down to 0° C., and then EDCI-HCl ((2.9 g, 15.1 mmol, 1.2 eq) was added in batches, and stirred at room temperature overnight. The acetonitrile was removed under reduced pressure, and the residue was extracted by EA (100 mL×3), washed with saturated saline (100 mL), dried with anhydrous sodium sulfate, and then concentrated and purified by normal-phase flash column chromatography (5% MeOH in CH2Cl2) to give white solid compound (6.6 g, 84.9%). MS (ESI): m/z found [M+1]+=648.3.
To a solution of compound 41 g (3.3 g, 5.1 mmol) in THF (30 mL) and H2O (20 mL) was added 10% Pd/C (1.0 g) and. After evacuation, the reaction was carried out under a hydrogen atmosphere for 20 h. The insoluble material was filtered off and concentrated to obtain the compound 41h (3.0 g, 68.6%) as a white solid. MS (ESI): m/z found [M+1]+=424.2.
To a solution of compound 41h (0.21 g, 0.5 mmol, 1.05 eq) in H2O (2.5 mL), a solution of compound 41i (0.41 g, 1.3 mmol, 1.1 eq) in ACN (2.5 mL) was added, followed by the addition of triethylamine (65 μL, 0.47 mmol, 1.0 eq), and the reaction was carried out with stirring at room temperature for 2 h. The reaction was purified by reversed-phase flash column chromatography (28% ACN in H2O, 0.5% HCOOH) to obtain the white solid compound 41 (MAL-PEG2-GGFG-OCH2COOH) (0.3 g, 87.0%). MS (ESI): m/z found [M−1]−=732.3.
Compound 41d (10.5 g, 25.4 mmol, 1.0 eq) was dissolved in tetrahydrofuran (100 mL). HOSu (3.22 g, 27.9 mmol, 1.1 eq) and DCC (5.76 g, 27.9 mmol, 1.1 eq) were added, and stirred for 2 h at room temperature. The filtrate was filtered and concentrated to give a semi-solid product which was used directly in the next step without purification. MS (ESI): m/z found [M+1]+=511.2.
Compound 42a (12.9 g, 25.4 mmol) and glycine (2.04 g, 27.9 mmol, 1.1 eq) were dissolved in acetonitrile (55 mL) and water (55 mL) with the addition of triethylamine (4.0 mL, 27.9 mmol, 1.1 eq), and stirred for 2 h at room temperature. The pH was adjusted to 2-3 with hydrochloric acid, and the reaction solution was extracted with ethyl acetate, washed with saturated brine, dried and concentrated to obtain a white foamy solid (11.9 g, 1000%). MS (ESI): m/z found [M+1]+=471.2.
Compound 42b (11.5 g, 24.5 mmol), p-aminobenzyl alcohol (4.51 g, 36.7 mmol, 1.5 eq) and EEDQ (9.07 g, 36.7 mmol, 1.5 eq) were dissolved in dichloromethane (160 mL) and methanol (80 mL) and stirred at room temperature overnight. The solvent was drained, and the resulting mixture was dissolved with ethyl acetate, washed with saturated saline, dried and concentrated to obtain a yellow solid (5.93 g, 42% c). MS (ESI): m/z found [M−17]+=558.2.
Compound 42d (5.93 g, 10.3 mmol) was dissolved in methanol (100 mL), and palladium carbon (3.0 g) was added, and stirred under hydrogen atmosphere for 2.5 h. TLC (EA) monitored the disappearance of the raw material. Filtered with diatomaceous earth and concentrated to obtain a white semi-solid (3.67 g, 81%). MS (ESI): m/z found [M+1]+=442.2.
1H NMR (400 MHz, CDCl3): δ 9.77 (s, 1H), 8.47 (t, J=6.0 Hz, 1H), 8.30 (d, J=8.0 Hz, 1H), 8.07 (s, 1H), 7.57 (d, J=8.4 Hz, 2H), 7.27-7.18 (m, 7H), 5.76 (br, 1H), 4.53-4.49 (m, 1H), 4.43 (s, 2H), 3.94-3.77 (m, 3H), 3.66 (d, J=16.4 Hz, 1H), 3.10-3.06 (m, 3H), 2.82 (dd, J=10.0 Hz, 13.8 Hz, 1H), 2.06 (br, 2H).
Compound 42e (1.64 g, 3.7 mmol, 1.0 eq) and compound 38a (2.35 g, 4.1 mmol, 1.1 eq) were dissolved in DMF (16 mL). HATU (2.35 g, 4.1 mmol, 1.1 eq) and DIEA (0.72 mL, 4.5 mmol, 1.2 eq) were added, and mixed at room temperature overnight. Purification by reversed-phase column chromatography (45% ACN in H2O) to obtain a white solid (3.0 g, 810%). MS (ESI): m/z found [M−17]+=984.1.
Compound 42f (1.0 g) was dissolved in diethylamine (20 mL) and tetrahydrofuran (40 mL) and stirred at room temperature for 4 h. The reaction solution turned into a gelatinous liquid. The solvent was drained to give a viscous solid which was used directly in the next step. MS (ESI): m/z found [M+1]+=777.4.
Compound 42f and EMCA (233 mg, 1.1 eq) were dissolved in DMF (5 mL). HATU (419 mg, 1.1 eq) and DIEA (179 μL, 1.1 eq) were added and stirred at room temperature overnight. Purification by reversed-phase column chromatography (36% ACN in H2O) to obtain a pink solid (607 mg, 58%). MS (ESI): m/z found [M−1]+=968.3.
Compound 43a (1.00 g, 2.99 mmol, 1.0 eq) was dissolved in and tetrahydrofuran (20 mL). Compound 43b (0.20 g, 3.64 mmol, 1.2 eq) and DIEA (0.74 mL, 1.5 eq) were added, and stirred for 3 h at room temperature. The reaction solution was purified by reversed-phase column chromatography to obtain the compound 43c. MS (ESI): m/z found [M+1]+=275.1.
Fmoc-L-valine (30.0 g, 88.5 mmol, 1.0 eq), L-alanine tert-butyl ester hydrochloride (17.68 g, 97.3 mmol, 1.1 eq), and HATU (40.35 g, 106.2 mmol, 1.2 eq) were dissolved in DMF (300 mL), and DIEA (32 mL, 0.19 μmol, 2.2 eq) was added and stirred at room temperature for 5 h. The reaction solution was poured into water to precipitate a white solid, filtered and dried to obtain the compound 43f (39.0 g, 99%). MS (ESI): m/z found [M−55]+=411.2.
Compound 43f (39.0 g, 83.7 mmol) was dissolved in CH2Cl2D (300 mL), and then TFA (100 mL) was added. after the reaction was reacted for 3 h. LC-MS monitoring showed that the reaction was complete. The reaction solution was poured into water, pulped 2 times, filtered, and the filter residue was the product. The product was placed in drying oven and dried to obtain the compound 43 g (30.9 g, 90%). MS (ESI): m/z found [M+1]+=411.2.
Compound 43g (5.0 g, 12.2 mmol, 1.0 eq), compound 42c (2.3 g, 18.7 mmol, 1.5 eq), HATU (5.1 g, 13.4 mmol, 1.1 eq), and DIEA (4.73 g, 36.6 mmol, 3.0 eq) were dissolved in DMF (30 mL). After the reaction was carried out for 3 h, LC-MS monitoring showed that the reaction was complete. The reaction solution was poured into water, pulped twice, filtered, and the filter residue was the product. The product was placed in an oven and dried to yield compound 43h (5.3 g, 84%). MS (ESI): m/z found [M+1]+=516.2.
Et2NH (36 mL) was added to a solution of compound 43 h (2.6 g, 5 mmol) in THF (140 mL). After the reaction was carried out for 3 h, LC-MS monitoring showed that the reaction was complete. The reaction solution was purified by column chromatography to obtain compound 7 (1.1 g, 73%). MS (ESI): m/z found [M+1]+=294.2.
1H NMR (400 MHz, DMSO): δ 10.03 (s, 1H), 8.28 (s, 1H), 7.53 (d, J=4 Hz, 2H), 7.25 (d, J=8.4 Hz, 2H), 5.12 (s, 1H), 4.48-4.43 (m, 1H), 4.42 (s, 2H), 3.12 (d, J=5.2 Hz, 1H), 1.97-1.92 (m, 3H), 1.31 (d, J=6.8 Hz, 3H), 1.31 (d, J=6.8 Hz, 3H) 3.12 (d, J=5.2 Hz, 1H), 1.97-1.92 (m, 1H), 1.31 (d, J=6.8 Hz, 31H), 0.89 (dd, J=33.6, 6.8 Hz, 2H) ppm.
Compound 43i (1.00 g, 3.41 mmol, 1.0 eq) was dissolved in tetrahydrofuran (10 mL), and diglycolic anhydride (0.44 g, 3.79 mmol, 1.1 eq) was added and stirred at room temperature for 2 h. The reaction solution was concentrated to obtain compound 43k, which was used directly in the next step. MS (ESI): m/z found [M−1]−=409.2.
Compound 43k (1.4 g, 3.42 mmol, 1.0 eq) was dissolved in tetrahydrofuran (20 mL). HOSu (0.47 g, 4.09 mmol, 1.2 eq) and DCC ((0.85 g, 4.13 mmol, 1.2 eq) were added, and stirred for 12 h at room temperature. The filtrate was filtered, concentrated and then purified by column chromatography (5% MeOH in CH2C2) to obtain compound 431. MS (ESI): m/z found [M+1]+=507.2.
1H NMR (400 MHz, CDCl3): δ 9.02 (s, 1H), 8.90 (s, 1H), 7.79 (s, 1H), 7.69 (t, J=6.8 Hz, 3H), 7.58 (d, J=8.4 Hz, 2H), 7.28 (s, 1H), 6.69 (s, 2H), 5.29 (s, 1H), 4.69-4.63 (m, 1H), 4.61 (s, 2H), 4.51-4.41 (m, 4H), 4.37-4.31 (m, 1H), 4.15 (d, J=14.0 Hz, 2H), 4.05-3.93 (m, 2H), 3.82 (t, J=4.9 Hz, 2H1), 3.67-3.49 (m, 30H1), 3.49-3.42 (m, 2H1), 3.34 (d, J=6.8 Hz, 2H), 3.10 (dd, J=14.7, 7.1 Hz, 1H), 2.68 (s, 1H), 2.29-2.20 (m, 1H), 1.85 (d, J=11.7 Hz, 2H), 1.70 (d, J=12.1 Hz, 2H), 1.56-1.39 (m, 8H), 0.98 (t, J=7.4 Hz, 6H).
Compound 431 (1.00 g, 1.98 mmol, 1.2 eq) and compound 43m (0.72 g, 1.64 mmol, 1.0 eq) were dissolved in tetrahydrofuran (20 mL), and DIEA (0.3 mL, 1.98 mmol, 1.0 eq) was added, and stirred for 3 h at room temperature. The reaction solution was concentrated and purified by silica gel column chromatography (5% MeOH in CH2Cl2) to obtain the compound 43n (11.9 g, 100%). MS (ESI): m/z found [M−1]−=828.4.
Compound 43c (223 mg, 0.81 mmol, 1.2 eq) and compound 43n (562 mg, 0.68 mmol, 1.0 eq) were dissolved in tetrahydrofuran (15 mL) with the addition of cuprous iodide (155 mg, 0.82 mmol, 1.2 eq) and DIEA (0.35 mL, 3.0 eq), and stirred at 65° C. for 3 hours. The reaction solution was filtered and concentrated and then purified by silica gel column chromatography (7% MeOH in CH2Cl2) to obtain the compound 43 (500 mg, 66%). MS (ESI): m/z found [(M−18)/2+1]+=543.
1H NMR (400 MHz, CDCl3): δ 9.02 (s, 1H), 8.90 (s, 1H), 7.79 (s, 1H), 7.69 (t, J=6.8 Hz, 3H), 7.58 (d, J=8.4 Hz, 2H), 7.28 (s, 1H), 6.69 (s, 2H), 5.29 (s, 1H), 4.69-4.63 (m, 1H), 4.61 (s, 2H), 4.51-4.41 (m, 4H), 4.37-4.31 (m, 1H), 4.15 (d, J=14.0 Hz, 2H), 4.05-3.93 (m, 2H), 3.82 (t, J=4.9 Hz, 2H), 3.67-3.49 (m, 30H), 3.49-3.42 (m, 2H), 3.34 (d, J=6.8 Hz, 2H), 3.10 (dd, J=14.7, 7.1 Hz, 1H), 2.68 (s, 1H), 2.29-2.20 (m, 1H), 1.85 (d, J=11.7 Hz, 2H), 1.70 (d, J=12.1 Hz, 2H), 1.56-1.39 (m, 8H), 0.98 (t, J=7.4 Hz, 6H).
Compound 37t (1.33 g, 4.32 mmol, 1.0 eq) was dissolved in and tetrahydrofuran (20 mL), and compound 43b (0.28 g, 5.09 mmol, 1.2 eq) and DIEA (1.4 mL, 3.0 eq) were added, and stirred for 3 h at room temperature. The reaction solution was concentrated and purified by column chromatography (5% MeOH in CH2Cl2) to obtain the compound 44a. MS (ESI): m/z found [M+1]+=249.1.
Compound 44a (120 mg, 0.48 mmol, 1.2 eq) and compound 43m (335 mg, 0.4 mmol, 1.0 eq) were dissolved in tetrahydrofuran (10 mL) with the addition of cuprous iodide (92 mg, 0.48 mmol, 1.2 eq) and DIEA (0.2 mL, 3.0 eq), and stirred at 65° C. for 3 hours. The reaction solution was filtered, concentrated and then purified by silica gel column chromatography (7% MeOH in CH2C2) to obtain compound 44 (500 mg, 66%). MS (ESI): m/z found [(M−18)/2+1]+=530.
To a solution of compound 45a (1.0 g, 5.92 mmol, 1.0 eq) in ACN (5 mL) was added HOSu (0.82 g, 7.13 mmol, 1.2 eq) and DCC (1.46 g, 7.09 mmol, 1.2 eq) in an ice bath, and the reaction was stirred at room temperature for 12 h. At the end of the reaction, the insoluble material was filtered out, and the reaction was concentrated to obtain the yellow oily compound (1.5 g, 96%). MS (ESI): m/z found [M+1]+=267.1.
Compound 45b (1.0 g, 3.76 mmol, 1.0 eq) was dissolved in and tetrahydrofuran (20 mL), compound 43b (0.25 g, 4.55 mmol, 1.2 eq) and DIEA (1.2 mL, 2.0 eq) were added, and the reaction was stirred for 3 h at room temperature. The reaction solution was concentrated and then purified by column chromatography (4% MeOH in CH2Cl2) to obtain the compound 45c. MS (ESI): m/z found [M+1]+=207.1.
Compound 45c (152 mg, 0.74 mmol, 1.2 eq) and compound 43m (510 mg, 0.62 mmol, 1.0 eq) were dissolved in tetrahydrofuran (10 mL) with the addition of cuprous iodide (140 mg, 0.74 mmol, 1.2 eq) and DIEA (0.30 mL, 3.0 eq), and stirred at 65° C. for 3 hours. The reaction solution was filtered and concentrated and then purified by silica gel column chromatography (7% MeOH in CH2Cl2) to obtain the compound 45 (325 mg, 51%). MS (ESI): m/z found [(M−18)/2+1]+=509.
Compound 43i (500 mg, 1.7 mmol, 1.0 eq), imidazole (174 mg, 2.6 mmol, 1.5 eq) and TBSCl (334 mg, 2.2 mmol, 1.3 eq) were dissolved in ACN (20 mL), and the reaction was completed as shown by LC-MS monitoring after 16 h of reaction. The reaction solution was purified by column chromatography to obtain the compound 46a (625 mg, 90%). MS (ESI): m/z found [M+1]+=408.3.
Compound 46a (757 mg, 1.9 mmol, 1.0 eq), compound 37q (996 mg, 2.0 mmol, 1.1 eq), HATU (1.06 g, 2.8 mmol, 1.5 eq) and DIEA (920 μL, 5.6 mmol, 3.0 eq) were dissolved in DMF (20 mL), and the reaction was carried out for 5 h. The LC-MS monitoring showed that the reaction was completed. The reaction solution was extracted with EA and saturated saline, and the organic phase was dried and concentrated to obtain the crude product. The crude product was purified by column chromatography to obtain the compound 46b (615 mg, 50%). MS (ESI): m/z found [M−TBS−17]+=745.
Et2NH (4 mL) was added to a solution of compound 46b (600 mg, 0.7 mmol) in THF (20 mL). After the reaction was carried out for 3 h, LC-MS monitoring showed that the reaction was completed. The reaction solution was drained and purified by reversed-phase column chromatography to obtain the compound 46c (403 mg, 90%). MS (ESI): m/z found [M+H]+=655.
Compound 37t (392 mg, 1.9 mmol, 1.5 eq), compound 46c (811 mg, 1.2 mmol, 1.0 eq), HATU (942 mg, 2.5 mmol, 2.0 eq), and DIEA (480 mg, 3.7 mmol, 3.0 eq) were dissolved in DMF (20 mL), and the reaction was carried out for 3 h. After 3 h, LC-MS monitoring showed that the reaction was completed. The reaction solution was extracted with EA and saturated saline, and the organic phase was dried and concentrated to obtain the crude product. The crude product was purified by column chromatography to obtain the compound 46d (260 mg, 25%). MS (ESI): m/z found [M-TBS-17]+=716.
TFA (0.3 mL) and H2O (0.2 mL) were added to a solution of compound 46d (260 mg, 0.3 mmol) in ACN (6 mL), and the reaction was completed by LC-MS monitoring after 10 min. The reaction solution was spun dry, dissolved in DMF and purified to obtain the compound 46 (82 mg, 36%) on a C18 column. MS (ESI): m/z found [M+H]+=734.
Compound 43a (120 mg, 0.4 mmol, 1.5 eq), compound 46c (157 mg, 0.2 mmol, 1.0 eq) and DIEA (40 μL, 0.2 mmol, 1.0 eq) were dissolved in ACN (2 mL) for 1 h. After 1 h of the reaction, the reaction was completed by LC-MS monitoring. The reaction solution was purified by column chromatography to obtain the compound 47a (188 mg, 90%). MS (ESI): m/z found [M-TBS-17]+=742.
TFA (0.1 mL) and H2O (0.1 mL) were added to ACN (2 mL) solution of compound 47a (188 mg, 0.2 mmol), and the reaction was completed by LC-MS monitoring after 10 min. The reaction solution was spun dry and dissolved in DMF and purified by C18 column to obtain the compound 47 (130 mg, 80%). MS (ESI): m/z found [M+H]+=760.
Compound 2 (30 mg, 44 μmol, 1.0 eq) and compound 37 (33 mg, 47 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (25 mg, 66 μmol, 1.5 eq) and DIEA (18 μL, 110 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (38% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (35 mg, 61%). MS (ESI): m/z found [M/2+1]+=655.2.
1H NMR (400 MHz, CDCl3): δ 8.79 (s, 1H), 8.55 (t, J=6.4 Hz, 1H), 7.65 (d, J=6.4 Hz, 1H), 7.62-7.58 (m, 4H), 7.44 (d, J=8.0 Hz, 1H), 7.36 (d, J=8.4 Hz, 2H), 7.32-7.30 (m, 2H), 6.98 (d, J=8.2 Hz, 1H), 6.70 (d, J=5.2 Hz, 1H), 6.69 (s, 1H), 6.61 (t, J=5.3 Hz, 1H), 5.73 (d, J=16.2 Hz, 1H), 5.55-5.53 (m, 1H), 5.38 (d, J=16.2 Hz, 1H), 5.29 (d, J=19.1 Hz, 1H), 5.17 (d, J=19.0 Hz, 1H), 4.80 (q, J=10.0 Hz, 1H), 4.69 (q, J=10.0 Hz, 1H), 4.27 (t, J=6.7 Hz, 1H), 4.13-3.92 (m, 6H), 3.81-3.69 (m, 2H), 3.68-3.59 (m, 12H), 3.55 (t, J=5.2 Hz, 2H), 3.49 (t, J=7.2 Hz, 2H), 3.43 (q, J=5.2 Hz, 2H), 3.21-3.15 (m, 1H), 3.10-3.05 (m, 1H), 2.53 (q, J=6.0 Hz, 2H), 2.39 (s, 3H), 2.35-2.30 (m, 2H), 2.18 (t, J=7.6 Hz, 2H), 1.95-1.87 (m, 2H), 1.67-1.54 (m, 2H), 1.38 (d, J=7.1 Hz, 3H), 1.33-1.26 (m, 4H), 1.20 (d, J=7.1 Hz, 6H), 1.05 (t, J=7.4 Hz, 3H).
Compound 4 (19 mg, 29 μmol, 1.0 eq) and compound 37 (22 mg, 29 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (17 mg, 44 μmol, 1.5 eq) and DIEA (12 μL, 73 μmol, 2.5 eq) were added and stirred at room temperature for 3 hours. The reaction solution was directly purified by reverse phase column chromatography (33% CH3CN in H2O, 0.05% HCOOH) to obtain a pale yellow solid (18 mg, 48%). MS(ESI): m/z found Fragment 1, Positive=591.2, Fragment 2, Positive=683.2
1H NMR (400 MHz, CDCl3): δ 8.15 (d, J=8.4 Hz, 1H), 7.74 (t, J=6.2 Hz, 1H), 7.6 (s, 1H), 7.56-7.52 (m, 2H), 7.46 (d, J=5.6 Hz, 1H), 7.36 (d, J=6.6 Hz, 1H), 6.68 (s, 2H), 6.50 (t, J=5.7 Hz, 1H), 5.71-5.56 (m, 3H), 5.26 (d, J=16.4 Hz, 1H), 5.08 (d, J=19.2 Hz, 1H), 4.67 (dd, J=8.6, 2.8 Hz, 1H), 4.35 (t, J=9.8 Hz, 1H), 4.24 (t, J=7.0 Hz, 1H), 4.19-4.12 (m, 5H), 3.93 (d, J=15.7 Hz, 1H), 3.73-3.66 (m, 3H), 3.63-3.57 (m, 12H), 3.53 (t, J=5.2 Hz, 2H), 3.48 (t, J=7.2 Hz, 2H), 3.41 (t, J=5.4 Hz, 2H), 3.37-3.32 (m, 1H), 3.23-3.15 (m, 1H), 3.12-3.04 (m, 1H), 2.47-2.25 (m, 8H), 2.15 (t, J=7.5 Hz, 2H), 2.03-1.96 (m, 3H), 1.66-1.54 (m, 4H), 1.34 (d, J=3.2 Hz, 3H), 1.33 (d, J=3.1 Hz, 3H), 1.31-1.24 (m, 3H), 1.18 (dd, J=25.2, 7.3 Hz, 1H), 1.05-0.99 (m, 6H).
Compound 2 (30 mg, 44 μmol, 1.0 eq) and compound 38 (37 mg, 44 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (25 mg, 66 μmol, 1.5 eq) and DIEA (18 μL, 110 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (37% CH3CN in H2O, 0.05% HCOOH) to obtain a pale yellow solid (27 mg, 43%). MS(ESI): m/z found Fragment 1, Positive=627.2, Fragment 2, Positive=771.2.
1H NMR (400 MHz, CDCl3): δ 8.79 (s, 1H), 8.53 (t, J=6.3 Hz, 1H), 7.67 (d, J=5.8 Hz, 1H), 7.60 (s, 1H), 7.58 (s, 1H), 7.56 (s, 1H), 7.55 (s, 1H), 7.52 (t, J=5.9 Hz, 1H), 7.41 (d, J=7.7 Hz, 1H), 7.34 (s, 1H), 7.32 (s, 1H), 7.28 (s, 1H), 6.90 (d, J=5.5 Hz, 1H), 6.67 (s, 2H), 6.57 (t, J=5.3 Hz, 1H), 5.71 (d, J=16.3 Hz, 1H), 5.53 (q, J=7.6 Hz, 1H), 5.36 (d, J=20.2 Hz, 1H), 5.26 (d, J=19.2 Hz, 1H), 5.15 (d, J=20.1 Hz, 1H), 4.77 (dd, J=6.7, 10.2 Hz, 1H), 4.63 (dd, J=6.2, 10.2 Hz, 1H), 4.25-4.18 (m, 1H), 4.09 (d, J=16.2 Hz, 1H), 4.05-3.99 (m, 2H), 3.90 (d, J=16.0 Hz, 1H), 3.80-3.73 (m, 2H), 3.70-3.57 (m, 22H), 3.53 (t, J=5.1 Hz, 2H), 3.47 (t, J=7.0 Hz, 2H), 3.41 (q, J=5.2 Hz, 2H), 3.15 (dt, J=5.3, 17.0 Hz, 1H), 3.05 (t, J=7.9 Hz, 1H), 2.56-2.41 (m, 2H), 2.37 (s, 3H), 2.33 (s, 1H), 2.30-2.24 (m, 1H), 2.15 (t, J=7.4 Hz, 2H), 1.65-1.52 (m, 4H), 1.37 (d, J=7.1 Hz, 3H), 1.33-1.23 (m, 4H), 1.19 (d, J=7.0 Hz, 3H), 1.16 (d, J=7.2 Hz, 3H), 1.02 (t, J=7.4 Hz, 3H).
Compound 4 (30 mg, 46 μmol, 1.0 eq) and compound 38 (39 mg, 46 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (26 mg, 70 μmol, 1.5 eq) and DIEA (19 μL, 116 μmol, 2.5 eq) were added, stirred at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (34% CH3CN in H2O, 0.05% HCOOH) to obtain a pale yellow solid (28 mg, 44%). MS(ESI): m/z found Fragment 1, Positive=591.2, Fragment 2, Positive=771.2.
1H NMR (400 MHz, CDCl3): δ 8.17 (d, J=8.4 Hz, 1H), 7.68 (t, J=7.8 Hz, 1H), 7.61-7.50 (m, 4H), 7.36 (d, J=6.6 Hz, 1H), 6.68 (s, 2H), 6.52 (t, J=5.7 Hz, 1H), 5.68 (t, J=16.2 Hz, 2H), 5.58 (q, J=6.4 Hz, 1H), 5.27 (d, J=16.4 Hz, 1H), 5.10 (d, J=19.4 Hz, 1H), 4.68 (dd, J=8.4, 3.0 Hz, 1H), 4.31 (t, J=10.0 Hz, 1H), 4.22 (t, J=7.1 Hz, 1H), 4.17-4.04 (m, 5H), 3.89 (d, J=15.6 Hz, 1H), 3.75-3.67 (m, 3H), 3.66-3.56 (m, 20H), 3.53 (t, J=5.2 Hz, 2H), 3.49 (t, J=7.2 Hz, 2H), 3.41 (t, J=5.3 Hz, 2H), 3.33 (q, J=8.8 Hz, 1H), 3.24-3.16 (m, 1H), 3.12-3.04 (m, 1H), 2.49-2.23 (m, 8H), 2.16 (t, J=7.5 Hz, 2H), 1.92-1.86 (m, 3H), 1.66-1.54 (m, 4H), 1.35 (t, J=7.7 Hz, 6H), 1.31-1.25 (m, 3H), 1.18 (dd, J=25.2, 7.3 Hz, 1H), 1.03 (t, J=7.2 Hz, 3H), 1.01 (t, J=5.1 Hz, 3H).
Compound 2 (28 mg, 41 μmol, 1.0 eq) and compound 39 (38 mg, 41 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (23 mg, 62 μmol, 1.5 eq) and DIEA (17 μL, 103 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (37% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (32 mg, 52%). MS(ESI): m/z found Fragment 1, Positive=627.2, Fragment 2, Positive=859.4.
1H NMR (400 MHz, CDCl3): δ 8.79 (s, 1H), 8.53 (t, J=6.3 Hz, 1H), 7.68 (d, J=5.7 Hz, 1H), 7.60 (s, 1H), 7.58 (s, 2H), 7.55-7.51 (m, 2H), 7.43 (d, J=8.0 Hz, 1H), 7.34 (s, 1H), 7.31 (s, 2H), 6.87 (d, J=8.6 Hz, 1H), 6.67 s, 2H), 6.47 (t, J=5.5 Hz, 1H), 5.72 (d, J=16.3 Hz, 1H), 5.52 (q, J=7.2 Hz, 1H), 5.34 (d, J=16.2 Hz, 1H), 5.26 (d, J=19.2 Hz, 1H), 5.11 (d, J=18.9 Hz, 1H), 4.77 (dd, J=6.5, 11.8 Hz, 1H), 4.62 (dd, J=6.2, 9.9 Hz, 1H), 4.20 (t, J=6.6 Hz, 1H), 4.08 (d, J=16.1 Hz, 1H), 4.04-3.96 (m, 2H), 3.89 (d, J=16.0 Hz, 1H), 3.78-3.71 (m, 2H), 3.68-3.56 (m, 30H), 3.52 (t, J=5.0 Hz, 2H), 3.47 (t, J=7.3 Hz, 2H), 3.41 (q, J=5.1 Hz, 2H), 3.17-3.10 (m, 1H), 3.06-2.98 (m, 1H), 2.56-2.49 (m, 1H), 2.46-2.40 (m, 1H), 2.36 (s, 3H), 2.28-2.25 (m, 2H), 2.15 (t, J=7.6 Hz, 2H), 1.66-1.53 (m, 4H), 1.36 (d, J=7.1 Hz, 3H), 1.33-1.24 (m, 4H), 1.20 (d, J=7.1 Hz, 3H), 1.15 (d, J=7.2 Hz, 3H), 1.01 (t, J=7.3 Hz, 3H).
Compound 4 (25 mg, 39 μmol, 1.0 eq) and compound 39 (36 mg, 39 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (22 mg, 58 μmol, 1.5 eq) and DIEA (16 μL, 97 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (3400 CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (17 mg, 300%). MS(ESI): m/z found Fragment 1, Positive=591.2, Fragment 2, Positive=859.4. 1H NMR (400 MHz, CDCl3): δ 8.17 (d, J=8.6 Hz, 1H), 7.67-7.62 (m, 2H), 7.62-7.50 (m, 4H), 7.36 (d, J=6.6 Hz, 1H), 6.68 (s, 2H), 6.52 (t, J=5.8 Hz, 1H), 5.71 (dd, J=15.1, 7.7 Hz, 2H), 5.58 (q, J=7.4 Hz, 1H), 5.27 (d, J=16.2 Hz, 1H), 5.12 (d, J=19.4 Hz, 1H), 4.68 (dd, J=8.3, 2.9 Hz, 1H), 4.28 (t, J=9.6 Hz, 1H), 4.23-4.15 (m, 2H), 4.13-4.04 (m, 4H), 3.88 (d, J=15.7 Hz, 1H), 3.74-3.57 (m, 30H), 3.53 (t, J=4.7 Hz, 2H), 3.49 (t, J=7.2 Hz, 2H), 3.42 (t, J=5.2 Hz, 2H), 3.33 (q, J=8.9 Hz, 1H), 3.25-3.17 (m, 1H), 3.12-0.99 (m, 6H).
Compound 44 (27 mg, 25 μmol, 1.0 eq) was dissolved in DMF (0.5 mL). NPC (9.1 mg, 30 μmol, 1.2 eq) and DIEA (8 μL, 50 μmol, 2.0 eq) were added, and stirred at room temperature for 5 hours. Compound 4 (16.0 mg, 25 μmol, 1.0 eq) was added to the reaction solution, and stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (43% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (15 mg, 37%). MS(ESI):m/z found [M/2+1]+=819.0.
Compound 45 (33 mg, 32 μmol, 1.0 eq) was dissolved in DMF (0.5 mL). NPC (11.6 mg, 38 μmol, 1.2 eq) and DIEA (10.5 μL, 64 μmol, 2.0 eq) were added, and stirred at room temperature for 5 hours. Compound 4 (20.6 mg, 32 μmol, 1.0 eq) was added to the reaction solution, and stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (38% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (14 mg, 28%). MS(ESI):m/z found [M/2+1]+=798.0.
Compound 2 (38 mg, 56 μmol, 1.0 eq) and compound 40 (43 mg, 56 μmol, 1.0 eq) were dissolved in DMF (1 mL), and HATU (32 mg, 83 μmol, 1.5 eq) and DIEA (23 μL, 139 μmol, 2.5 eq) were added, and stirred at room temperature for 3 hours. Purified by HPLC to obtain a white solid (14 mg, 19%). MS(ESI):m/z found [M/2+1]+=663.6.
1H NMR (400 MHz, CDCl3): δ 9.44 (s, 1H), 8.15 (t, J=5.2 Hz, 1H), 7.64 (d, J=7.7 Hz, 1H), 7.60 (s, 2H), 7.56-7.54 (m, 2H), 7.53 (s, 1H), 7.48 (d, J=5.2 Hz, 1H), 7.35 (s, 1H), 7.33 (s, 1H), 7.19 (d, J=7.7 Hz, 1H), 6.79 (s, 1H), 6.67 (s, 2H), 6.44 (t, J=6.0 Hz, 1H), 5.72 (d, J=16.2 Hz, 1H), 5.67-5.61 (m, 1H), 5.33 (d, J=16.2 Hz, 1H), 5.31 (d, J=19.0 Hz, 1H), 5.12 (d, J=19.0 Hz, 1H), 4.42 (dd, J=6.6, 13.5 Hz, 1H), 4.26 (dd, J=6.0, 13.6 Hz, 1H), 4.15 (t, J=6.3 Hz, 2H), 4.05 (t, J=6.3 Hz, 2H), 3.81-3.73 (m, 2H), 3.71-3.57 (m, 15H), 3.52 (t, J=5.1 Hz, 2H), 3.46 (t, J=7.2 Hz, 2H), 3.39 (q, J=7.2 Hz, 2H), 3.23-3.18 (m, 1H), 3.10-3.05 (m, 1H), 3.44 (q, J=5.4 Hz, 2H), 3.36 (s, 3H), 2.29-2.22 (m, 1H), 2.13 (t, J=7.5 Hz, 2H), 1.91-1.86 (m, 3H), 1.62-1.53 (m, 4H), 1.33 (s, 3H), 1.31 (s, 3H), 1.29-1.25 (m, 2H), 1.11 (d, J=7.2 Hz, 3H), 1.01 (t, J=7.4 Hz, 3H)
Compound 46 (31 mg, 42 μmol, 1.0 eq) was dissolved in DMF (0.5 mL), NPC (15.2 mg, 50 μmol, 1.2 eq) and DIEA (14 μL, 84 μmol, 2.0 eq) were added, and stirred at room temperature for 5 hours. Compound 4 (27 mg, 42 μmol, 1.0 eq) was added to the reaction solution, and stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (41% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (27 mg, 500%). MS(ESI):m/z found [M/2+1]+=646.8.
1H NMR (400 MHz, DMSO-d6): δ 9.86 (d, J=11.3 Hz, 1H), 8.60 (t, J 9.2 Hz, 1H), 8.41 (d, J=7.5 Hz, 1H), 7.99 (d, J=8.0 Hz, 1H), 7.79 (m, 2H), 7.64 (dd, J 17.7, 8.3 Hz, 2H), 7.28 m, 4H), 6.51 (s, 1H), 5.47-5.37 (m, 2H), 5.19 (d, J=4.6 Hz, 1H), 5.07-4.91 (m, 3H), 4.48-4.36 (i, 1H), 4.20 (q, J=7.3, 5.4 Hz, 1H), 4.12 (t, J 7.7 Hz, 1H), 4.04 (dd, J=8.9, 5.1 Hz, 1H), 3.62-3.53 (m, 4H), 3.45 (i, 14H), 3.15 (q, J=6.0 Hz, 4H), 2.92 (d, J=9.8 Hz, 1H), 2.80-2.71 (m, 1H), 2.45 (d, J=5.9 Hz, 2H), 2.41-2.30 (m, 4H), 2.18-2.09 (m, 1H), 2.01 (t, J=7.4 Hz, 2H), 1.96-1.89 (i, 3H), 1.82 (dt, J=14.1, 7.5 Hz, 4H), 1.48-1.39 (m, 4H), 1.30 (t, J=7.0 Hz, 3H), 1.20-1.11 (m, 2H), 0.96 (t, J=7.1 Hz, 3H), 0.85 (t, J=7.0 Hz, 6H).
Compound 2 (30 mg, 44 μmol, 1.0 eq) and compound 41 (32 mg, 44 μmol, 1.0 eq) were dissolved in DMF (1 mL), and HATU (20.6 mg, 53 μmol, 1.2 eq) and DIEA (16 μL, 97 μmol, 2.2 eq), and stirred at room temperature for 2 hours. Purified by HPLC to obtain a white solid (28 mg, 48%). MS(ESI):m/z found [M/2+1]+=642.8.
1H NMR (400 MHz, DMSO-d6): δ 9.71 (s, 1H), 8.74 (t, J=6.8 Hz, 1H), 8.66 (d, J=8.6 Hz, 1H), 8.38 (t, J=5.7 Hz, 1H), 8.16 (t, J=9.1 Hz, 2H), 8.02 (q, J=6.7 Hz, 2H), 7.80 (d, J=10.9 Hz, 1H), 7.59 (d, J=8.3 Hz, 2H), 7.30 (s, 1H), 7.25-7.22 (m, 5H), 7.20 (s, 1H), 7.18-7.13 (m, 1H), 6.99 (s, 2H), 6.53 (s, 1H), 5.53 (dt, J=9.1, 4.8 Hz, 1H), 5.43 (s, 2H), 5.25 (d, J=18.8 Hz, 1H), 5.16 (d, J=18.9 Hz, 1H), 4.66 (d, J=6.5 Hz, 2H), 4.52-4.47 (m, 1H), 4.03 (s, 2H), 3.80-3.66 (m, 6H), 3.59 (m, 5H), 3.55 (s, 1H), 3.45 (s, 7H), 3.19-3.15 (m, 2H), 3.12 (q, J=5.7 Hz, 2H), 3.06 (dd, J=13.6, 4.3 Hz, 1H), 2.81 (dd, J=13.8, 9.6 Hz, 1H), 2.40 (s, 3H), 2.38 (t, J=6.5 Hz, 2H), 2.31 (t, J=7.2 Hz, 2H), 2.13-2.09 (m, 1H), 1.86 (tq, J=14.1, 7.1 Hz, 2H), 0.87 (t, J=7.3 Hz, 3H).
Compound 41 (23 mg, 31 μmol, 1.0 eq) was dissolved in DMF (1 mL). EDCI HCl (7 mg, 37 μmol, 1.2 eq) and HOSu (4.3 mg, 37 μmol, 1.2 eq) were added, and stirred at room temperature for 4 hours. Triethylamine (9 μL, 68 μmol, 2.2 eq) and compound 4 (20 mg, 31 μmol, 1.0 eq) were added to the reaction solution, and stirred at room temperature for 1.5 hours. Purified by reverse phase column chromatography (37% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (23 mg, 58%). MS(ESI):m/z found [M/2+1]+=643.0.
1H NMR (400 MHz, DMSO-d6): δ 8.48 (m, 2H), 8.29 (t, J=5.8 Hz, 1H), 8.21-8.11 (m, 2H), 8.06-7.98 (m, 2H), 7.78 (d, J=11.0 Hz, 1H), 7.30 (s, 1H), 7.26-7.17 (m, 5H), 7.00 (s, 2H), 6.53 (s, 1H), 5.53 (dt, J=9.3, 5.1 Hz, 1H), 5.43 (s, 2H), 5.28-5.17 (m, 2H), 4.48 (td, J=8.9, 4.5 Hz, 1H), 4.30 (d, J=6.5 Hz, 2H), 4.01 (s, 2H), 3.77-3.66 (m, 4H), 3.59 (m, 7H), 3.46 (s, 6H), 3.15 (m, 5H), 3.04 (dd, J=13.8, 4.4 Hz, 1H), 2.78 (dd, J=13.9, 9.7 Hz, 1H), 2.43-2.37 (m, 6H), 2.32 (t, J=7.3 Hz, 2H), 2.19-2.05 (m, 3H), 1.88 (td, J=14.2, 5.8 Hz, 5H), 0.88 (t, J=7.3 Hz, 3H).
Compound 47 (30 mg, 39 μmol, 1.0 eq) was dissolved in DMF (0.5 mL). NPC (14.4 mg, 47 μmol, 1.2 eq) and DIEA (13 μL, 79 μmol, 2.0 eq) were added, and stirred at room temperature for 5 hours. Compound 4 (25.5 mg, 39 μmol, 1.0 eq) was added to the reaction solution, then stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (45% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (20 mg, 38%). MS (ESI): m/z found [M/2+1]+=660.0.
Compound 42 (50 mg, 52 μmol, 1.0 eq) was dissolved in DMF (0.5 mL). NPC (19 mg, 62 μmol, 1.2 eq) and DIEA (17 μL, 103 μmol, 2.0 eq) were added, and stirred at room temperature for 4 hours. Compound 4 (33 mg, 52 μmol, 1.0 eq) was added to the reaction solution, and stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (41% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (25 mg, 32%). MS(ESI):m/z found [M/2+1]+=765.0.
1H NMR (400 MHz, DMSO-d6): δ 9.92 (m, 1H), 8.62-8.57 (m, 1H), 8.42-8.38 (m, 1H), 8.19-8.13 (m, 2H), 8.05-8.00 (m, 1H), 7.81-7.75 (m, 2H), 7.67-7.61 (m, 2H), 7.31-7.16 (m, 8H), 7.00 (s, 1H), 6.52 (s, 1H), 4.52-4.48 (m, 1H), 4.21-4.16 (m, 1H), 3.95-3.85 (m, 2H), 3.80-3.72 (m, 3H), 3.70-3.65 (m, 4H), 3.63-3.23 (m, 31H), 3.20-3.10 (m, 4H), 3.10-3.04 (m, 2H), 2.98-2.92 (m, 1H), 2.87-2.78 (m, 1H), 2.41-2.36 (m, 5H), 2.18-2.09 (m, 2H), 2.03 (t, J=7.6 Hz, 2H), 2.00-1.82 (m, 7H), 1.84 (t, J=6.4 Hz, 3H), 1.48-1.44 (m, 2H), 0.84 (t, J=7.2 Hz, 3H).
Compound 43 (20 mg, 18 μmol, 1.0 eq) was dissolved in DMF (0.5 mL). NPC (7 mg, 22 μmol, 1.2 eq) and DIEA (6 μL, 36 μmol, 2.0 eq) were added, and stirred at room temperature for 5 hours. Compound 4 (12 mg, 18 μmol, 1.0 eq) was added to the reaction solution, and stirring was continued at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (41% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (9 mg, 30%). MS(ESI):m/z found [M/2+1]+=832.0.
Compound 24 (9 mg, 13 μmol, 1.0 eq) and compound 41 (9.6 mg, 13 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (6.0 mg, 16 μmol, 1.2 eq) and DIEA (5 μL, 33 μmol, 2.5 eq) were added, and stirred for 3 hours at room temperature. The reaction solution was directly purified by reversed-phase column chromatography (41% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (14 mg, 83%). MS(ESI): m/z found Fragment 1, Positive=627.2, Fragment 2, Positive=658.2.
1H NMR (400 MHz, DMSO-d6): δ 10.14 (s, 1H), 8.86 (d, J=8.7 Hz, 1H), 8.67 (t, J=6.7 Hz, 1H), 8.35 (t, J=5.9 Hz, 1H), 8.21-8.09 (m, 2H), 8.00 (t, J=5.7 Hz, 2H), 7.80 (d, J=10.9 Hz, 1H), 7.65 (d, J=8.0 Hz, 1H), 7.30 (s, 1H), 7.27-7.18 (m, 6H), 7.15 (dt, J=6.0, 2.8 Hz, 1H), 7.10 (t, J=7.4 Hz, 1H), 6.97 (s, 2H), 6.52 (s, 1H), 5.53 (dt, J=9.0, 4.6 Hz, 1H), 5.42 (s, 2H), 5.26 (d, J=18.9 Hz, 1H), 5.12 (d, J=19.0 Hz, 1H), 4.72 (d, J=6.7 Hz, 2H), 4.48 (td, J=8.6, 4.3 Hz, 1H), 4.07 (s, 2H), 3.75 (t, J=5.6 Hz, 2H), 3.67 (d, J=5.6 Hz, 2H), 3.60-3.53 (m, 6H), 3.43 (s, 4H), 3.31 (s, 2H), 3.18-3.09 (m, 4H), 3.03 (dd, J=13.8, 4.4 Hz, 1H), 2.77 (dd, J=13.8, 9.7 Hz, 1H), 2.40 (s, 3H), 2.36 (t, J=6.5 Hz, 2H), 2.30 (t, J=7.3 Hz, 2H), 2.19-2.03 (m, 2H), 1.90-1.79 (m, 2H), 0.85 (t, J=7.3 Hz, 3H).
Compound 21 (12 mg, 17 μmol, 1.0 eq) and compound 41 (19 mg, 26 μmol, 1.5 eq) were dissolved in DMF (0.5 mL). HATU (9.8 mg, 26 μmol, 1.5 eq) and DIEA (7 μL, 43 μmol, 2.5 eq) were added, and stirred at 50° C. for 3 hours. The reaction solution was directly purified by reverse phase column chromatography (40% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (14 mg, 83%). MS(ESI):m/z found [M/2+1]+=615.8.
Compound 19 (11.5 mg, 16 μmol, 1.0 eq) and compound 41 (18 mg, 24 μmol, 1.5 eq) were dissolved in DMF (0.5 mL). HATU (9.2 mg, 24 μmol, 1.5 eq) and DIEA (7 μL, 43 μmol, 2.5 eq) were added, and stirred at room temperature for 3 hours. The reaction solution was directly purified by reversed-phase column chromatography (40% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (12 mg, 57%). MS(ESI):m/z found [M/2+1]=657.2.
1H NMR (400 MHz, DMSO-d6): δ 9.73 (s, 1H), 8.80 (t, J=6.6 Hz, 1H), 8.56 (d, J=8.7 Hz, 1H), 8.48 (s, 1H), 8.44 (t, J=5.8 Hz, 1H), 8.27-8.18 (m, 2H), 8.08 (t, J=5.7 Hz, 1H), 8.04 (t, J=5.7 Hz, 1H), 7.81 (d, J=11.0 Hz, 1H), 7.39 (d, J=2.0 Hz, 1H), 7.33 (s, 1H), 7.27-7.21 (m, 5H), 7.18 (dq, J=5.8, 2.9 Hz, 1H), 7.11 (d, J=8.3 Hz, 1H), 7.00 (s, 1H), 6.56 (s, 1H), 5.55 (dt, J=9.0, 4.8 Hz, 1H), 5.44 (s, 2H), 5.23 (q, J=19.0 Hz, 2H), 4.68 (d, J=6.7 Hz, 2H), 4.50 (dt, J=8.7, 4.3 Hz, 1H), 4.05 (s, 2H), 3.80-3.73 (m, 2H), 3.70 (d, J=5.5 Hz, 2H), 3.65 (s, 2H), 3.61-3.56 (m, 6H), 3.50-3.45 (m, 5H), 3.19 (t, J=6.5 Hz, 2H), 3.14 (t, J=5.7 Hz, 2H), 3.11-3.04 (m, 2H), 2.44-2.36 (m, 5H), 2.32 (t, J=7.3 Hz, 2H), 2.15 (tt, J=12.9, 7.5 Hz, 2H), 2.04-1.97 (m, 1H), 1.87 (dp, J=21.4, 7.1 Hz, 3H), 1.24 (d, J=3.2 Hz, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 29 (18 mg, 27 μmol, 1.0 eq) and compound 38 (23 mg, 27 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (12 mg, 33 μmol, 1.2 eq) and DIEA (11 μL, 68 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (40% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (18 mg, 48%). MS(ESI): m/z found Fragment 1, Positive=605.2 Fragment 2, Positive=711.4.
1H NMR (400 MHz, DMSO-d6): δ 8.60 (t, J=6.6 Hz, 1H), 8.45 (s, 1H), 8.15 (dd, J=15.4, 6.9 Hz, 2H), 8.03 (d, J=7.3 Hz, 1H), 7.83 (t, J=5.7 Hz, 1H), 7.79 (d, J=10.6 Hz, 1H), 7.31 (d, J=6.7 Hz, 1H), 7.01 (s, 2H), 6.54 (s, 1H), 5.61 (q, J=6.5 Hz, 1H), 5.43 (s, 2H), 5.18-5.09 (m, 2H), 4.61-4.47 (m, 2H), 4.22-4.16 (m, 4H), 4.08 (td, J=14.4, 7.0 Hz, 2H), 3.61-3.54 (m, 4H), 3.49 (dd, J=8.5, 4.8 Hz, 23H), 3.23-3.11 (m, 6H), 2.40-2.36 (m, 5H), 2.33 (q, J=8.1, 7.2 Hz, 1H), 2.21 (d, J=13.4 Hz, 1H), 2.16 (q, J=6.9 Hz, 2H), 2.03 (t, J=7.4 Hz, 2H), 1.87 (dp, J=21.3, 7.1 Hz, 2H), 1.63 (d, J=12.2 Hz, 2H), 1.20-1.14 (m, 6H), 1.17 (m, 11H), 0.88 (t, J=7.3 Hz, 3H).
Compound 30 (22 mg, 33 μmol, 1.0 eq) and compound 38 (28 mg, 33 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (15 mg, 40 μmol, 1.2 eq) and DIEA (14 μL, 83 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reversed-phase column chromatography (40% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (19 mg, 42%). MS(ESI): m/z found Fragment 1, Positive=605.2 Fragment 2, Positive=711.4.
Compound 36 (19 mg, 29 μmol, 1.0 eq) and compound 38 (25 mg, 29 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (13 mg, 35 μmol, 1.2 eq) and DIEA (12 μL, 73 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (38% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (18 mg, 47%). MS(ESI): m/z found Fragment 1, Positive=519.2 Fragment 2, Positive=711.4.
1H NMR (400 MHz, DMSO-d6): δ 8.62 (t, J=6.8 Hz, 1H), 8.48 (d, J=8.9 Hz, 1H), 8.13 (dd, J=7.0, 4.8 Hz, 2H), 8.00 (d, J=7.3 Hz, 1H), 7.86-7.78 (m, 2H), 7.32 (s, 1H), 7.01 (s, 2H), 6.54 (s, 1H), 5.58 (dt, J=9.0, 4.6 Hz, 1H), 5.43 (s, 2H), 5.22 (q, J=18.8 Hz, 2H), 4.56-4.47 (m, 2H), 4.24 (dd, J=8.4, 4.4 Hz, 1H), 4.19 (pd, J=7.2, 2.8 Hz, 2H), 4.06 (qd, J=14.7, 7.9 Hz, 3H), 3.60-3.54 (m, 2H), 3.52-3.44 (m, 22H), 3.39-3.36 (m, 6H), 3.17 (q, J=5.9 Hz, 3H), 2.40-2.36 (m, 5H), 2.36-2.30 (m, 1H), 2.13-2.06 (m, 2H), 2.03 (t, J=7.4 Hz, 2H), 1.93 (td, J=11.0, 5.2 Hz, 1H), 1.87 (dq, J=10.3, 6.8 Hz, 3H), 1.47 (q, J=7.5 Hz, 4H), 1.17 (q, J=6.8 Hz, 9H), 1.12 (d, J=7.3 Hz, 3H), 0.91-0.86 (m, 3H).
Compound 19 (27 mg, 38 μmol, 1.0 eq) and compound 38 (32 mg, 38 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (24 mg, 46 μmol, 1.2 eq) and DIEA (16 μL, 95 μmol, 2.5 eq) were added, and stirred at room temperature for 3 hours. The reaction solution was directly purified by reversed-phase column chromatography (37% CH3CN in H2O, 0.05% HCOOH) to obtain a yellow solid (18 mg, 33%). MS(ESI):m/z found [M/2+1]=714.2.
1H NMR (400 MHz, DMSO-d6): δ 9.65 (s, 1H), 8.76 (t, J=6.6 Hz, 1H), 8.52 (d, J=8.6 Hz, 1H), 8.16-8.07 (m, 3H), 7.82-7.77 (m, 2H), 7.35 (d, J=1.9 Hz, 1H), 7.30 (s, 1H), 7.19 (dd, J=8.1, 1.9 Hz, 1H), 7.10 (d, J=8.2 Hz, 1H), 6.98 (s, 2H), 6.53 (s, 1H), 5.54 (dt, J=9.3, 4.8 Hz, 1H), 5.42 (s, 2H), 5.26 (d, J=18.8 Hz, 1H), 5.17 (d, J=18.8 Hz, 1H), 4.69-4.61 (m, 2H), 4.20 (ddd, J=9.0, 4.9, 2.1 Hz, 3H), 4.01 (s, 2H), 3.64 (s, 3H), 3.60-3.53 (m, 2H), 3.50-3.43 (m, 20H), 3.40 (s, 2H), 3.36-3.31 (m, 4H), 3.15 (dt, J=11.7, 6.9 Hz, 4H), 2.40 (d, J=1.8 Hz, 3H), 2.38-2.29 (m, 2H), 2.19-2.08 (m, 2H), 2.01 (t, J=7.4 Hz, 2H), 1.86 (ddd, J=32.5, 14.1, 7.2 Hz, 2H), 1.44 (p, J=7.4 Hz, 4H), 1.23 (d, J=7.2 Hz, 3H), 1.20-1.12 (m, 8H), 0.86 (t, J=7.3 Hz, 3H).
Compound 21 (23 mg, 33 μmol, 1.0 eq) and compound 38 (28 mg, 33 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (15 mg, 39 μmol, 1.2 eq) and DIEA (145 μL, 8315 μmol, 2.5 eq) were added, and stirred at room temperature for 3 hours. The reaction solution was directly purified by reverse phase column chromatography (37% CH3CN in H2O, 0.0500 HCOOH) to obtain a white solid (24 mg, 520%). MS(ESI): m/z found Fragment 1, Positive=645.0 Fragment 2, Positive=771.2.
1H NMR (400 MHz, DMSO-d6): δ 9.91 (s, 1H), 8.80 (t, J=6.6 Hz, 1H), 8.72 (d, J=8.5 Hz, 1H), 8.21-8.10 (m, 3H), 7.85-7.81 (m, 2H), 7.61 (dd, J=12.3, 2.0 Hz, 1H), 7.39 (dd, J=8.4, 2.0 Hz, 1H), 7.32 (s, 1H), 7.29 (d, J=8.5 Hz, 1H), 7.01 (s, 2H), 6.55 (s, 1H), 5.56 (dt, J=8.8, 4.6 Hz, 1H), 5.44 (s, 2H), 5.29 (d, J=19.0 Hz, 1H), 5.17 (d, J=19.0 Hz, 1H), 4.72-4.62 (m, 2H), 4.22 (td, J=7.1, 2.8 Hz, 3H), 4.05 (s, 2H), 3.60-3.54 (m, 3H), 3.52-3.44 (m, 21H), 3.37 (dd, J=6.8, 2.5 Hz, 4H), 3.21-3.14 (m, 4H), 2.42 (d, J=1.9 Hz, 4H), 2.42 (s, 3H), 2.39-2.31 (m, 2H), 2.23-2.08 (m, 2H), 2.03 (d, J=7.4 Hz, 2H), 1.86 (dq, J=14.1, 7.0 Hz, 2H), 1.46 (p, J=7.4 Hz, 4H), 1.29-1.12 (m, 11H), 0.88 (t, J=7.3 Hz, 3H).
R-Exatecan 71b (28.6 mg, 54 μmol, 1 eq) and N-Boc-L-proline 71a (12 mg, 56 μmol, 1.05 eq) were dissolved in DMF (1 mL). HATU (25 mg, 65 μmol, 1.2 eq) and DIEA (22 μL, 134 μmol, 2.5 eq) were added, and stirred at room temperature for 1.5 hours. The reaction solution changed from cloudy to clear. Purified by reverse phase column chromatography (50% ACN in H2O, HCOOH) obtain a white solid (34.5 mg, 100%). MS (ESI): m/z found [M+1]+=633.0.
Compound 71c (34.5 mg, 54 μmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (2 mL), and stirred at room temperature for 2.5 hours. Concentrated and then purified by reverse phase column chromatography (34% CH3CN in H2O, 0.1% TFA) obtain a yellow solid (18.2 mg, 52%). MS (ESI): m/z found [M+1]+=533.0.
1H NMR (600 MHz, DMSO-d6): δ 9.11 (br, 1H), 9.00 (d, J=8.4 Hz, 1H), 8.73 (br, 1H), 7.86 (d, J=10.9 Hz, 1H), 7.34 (s, 1H), 6.56 (s, 1H), 5.63 (dt, J=8.0, 3.7 Hz, 1H), 5.48-5.41 (m, 2H), 5.40 (d, J=15.6 Hz, 1H), 5.27 (d, J=18.9 Hz, 1H), 4.14-4.10 (m, 1H), 3.30-3.26 (m, 1H), 3.25-3.19 (m, 2H), 3.12 (dt, J=17.3, 8.5 Hz, 1H), 2.43 (s, 4H), 2.29-2.21 (m, 2H), 2.12 (tt, J=13.1, 4.6 Hz, 1H), 1.90 (m, 5H), 0.88 (t, J=7.3 Hz, 3H).
Compound 71 (16 mg, 25 μmol, 1.0 eq) and compound 38 (24 mg, 28 μmol, 1.0 eq) were dissolved in DMF (0.5 mL). HATU (14.5 mg, 38 μmol, 1.5 eq) and DIEA (10 μL, 64 μmol, 2.5 eq) were added, and stirred at room temperature for 2 hours. The reaction solution was directly purified by reverse phase column chromatography (42% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (18 mg, 520%). MS (ESI): m/z found Fragment 1, Positive=591.0, Fragment 2, Positive=771.2.
1H NMR (400 MHz, DMSO-d6) δ: 8.60 (t, J=6.8 Hz, 1H), 8.51 (d, J=8.8 Hz, 1H), 8.15 (d, J=7.3 Hz, 1H), 8.12 (d, J=6.5 Hz, 1H), 8.01 (d, J=7.1 Hz, 1H), 7.85-7.78 (m, 2H), 7.32 (s, 1H), 7.01 (s, 2H), 6.53 (s, 1H), 5.58 (dt, J=9.3, 4.7 Hz, 1H), 5.46-5.40 (m, 2H), 5.26-5.16 (m, 2H), 4.58-4.53 (m, 2H), 4.24 (dd, J=5.5, 2.9 Hz, 1H), 4.20 (tt, J=7.0, 3.1 Hz, 2H), 4.09 (q, J=7.1 Hz, 1H), 4.05 (s, 2H), 3.60-3.54 (m, 3H), 3.53-3.44 (m, 21H), 3.39-3.35 (m, 6H), 3.17 (q, J=5.9 Hz, 3H), 2.42-2.36 (m, 4H), 2.33 (dt, J=14.6, 6.2 Hz, 1H), 2.13-2.06 (m, 3H), 2.02 (t, J=4.9, 2H), 1.97-1.79 (m, 4H), 1.46 (p, J=7.4 Hz, 4H), 1.23-1.13 (m, 9H), 1.11 (d, J=7.2 Hz, 2H), 0.87 (t, J=7.3 Hz, 3H).
Compound 2 (20 mg, 29 μmol, 1.0 eq) and compound McGGFG (20 mg, 32 μmol, 1.1 eq) were dissolved in DMF (0.5 mL). HATU (11 mg, 35 μmol, 1.2 eq) and DIEA (12 μL, 11 μmol, 2.2 eq) were added, and stirred at room temperature for 3 hours. The reaction solution was directly purified by reversed-phase column chromatography (42% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (9.8 mg, 29%). MS (ESI): m/z found Fragment 1, Positive=541.0 Fragment 2, Positive=627.0.
1H NMR (400 MHz, DMSO-d6): δ 9.71 (d, J=4.0 Hz, 1H), 8.74 (t, J=6.7 Hz, 1H), 8.70-8.65 (m, 1H), 8.37 (t, J=5.8 Hz, 1H), 8.17 (d, J=8.2 Hz, 1H), 8.08 (t, J=5.8 Hz, 1H), 8.03 (t, J=5.9 Hz, 1H), 7.82 (d, J=10.9 Hz, 1H), 7.60 (d, J=8.7 Hz, 2H), 7.32 (d, J=2.9 Hz, 1H), 7.28-7.21 (m, 6H), 7.18 (ddt, J=7.3, 5.5, 2.4 Hz, 1H), 6.99 (s, 2H), 6.54 (d, J=2.3 Hz, 1H), 5.54 (dd, J=9.1, 4.5 Hz, 1H), 5.44 (d, J=5.5 Hz, 2H), 5.26 (dd, J=18.7, 4.9 Hz, 1H), 5.16 (dd, J=18.6, 5.0 Hz, 1H), 4.67 (d, J=6.7 Hz, 2H), 4.51 (td, J=8.8, 4.4 Hz, 1H), 4.05 (s, 2H), 3.81-3.72 (m, 3H), 3.67 (d, J=5.5 Hz, 2H), 3.60 (dd, J=16.6, 5.4 Hz, 1H), 3.46 (t, J=4.4 Hz, 2H), 3.37 (t, J=7.1 Hz, 2H), 3.17 (d, J=6.4 Hz, 2H), 3.07 (dd, J=13.9, 4.6 Hz, 1H), 2.82 (dd, J=13.9, 9.6 Hz, 1H), 2.41 (s, 3H), 2.17-2.07 (m, 4H), 1.87 (ddp, J=21.4, 14.4, 7.3 Hz, 2H), 1.46 (ddd, J=19.0, 11.2, 6.3 Hz, 4H), 1.19 (tt, J=9.8, 6.5 Hz, 2H), 0.88 (td, J=7.4, 3.2 Hz, 3H).
Compound 4 (25 mg, 39 μmol, 1.0 eq) and compound McGGFG (26 mg, 43 μmol, 1.1 eq) were dissolved in DMF (0.5 mL), and HATU (18 mg, 46 μmol, 1.2 eq) and DIEA (14 μL, 85 μmol, 2.2 eq), and stirred at room temperature for 3 hours. The reaction solution was directly purified by reverse phase column chromatography (38% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (10.3 mg, 23%). MS (ESI): m/z found Fragment 1, Positive=541.0 Fragment 2, Positive=591.0.
1H NMR (600 MHz, DMSO-d6) δ 8.50-8.44 (m, 2H), 8.27 (t, J=5.9 Hz, 1H), 8.14 (d, J=8.1 Hz, 1H), 8.08 (t, J=5.8 Hz, 1H), 8.01 (t, J=5.8 Hz, 1H), 7.78 (d, J=10.9 Hz, 1H), 7.31 (s, 1H), 7.24 (dt, J=13.4, 7.4 Hz, 4H), 7.19 (q, J=7.5, 6.7 Hz, 1H), 7.00 (s, 2H), 6.53 (d, J=6.9 Hz, 1H), 5.53 (dt, J=9.4, 5.1 Hz, 1H), 5.43 (d, J=5.3 Hz, 2H), 5.29-5.17 (m, 2H). 4.48 (td, J=8.9, 4.5 Hz, 1H), 4.31 (td, J=7.6, 6.0, 3.3 Hz, 2H), 4.01 (s, 2H), 3.73 (dd, J=16.6, 5.9 Hz, 1H), 3.66 (d, J=5.7 Hz, 2H), 3.59 (dd, J=16.4, 5.6 Hz, 2H), 3.53 (dd, J=16.5, 5.7 Hz, 1H), 3.44 (dt, J=12.7, 5.7 Hz, 1H), 3.36 (t, J=7.2 Hz, 3H), 3.17 (t, J=7.4 Hz, 2H), 3.04 (dd, J=13.9, 4.6 Hz, 1H), 2.79 (dd, J=13.8, 9.6 Hz, 1H), 2.40 (d, J=6.7 Hz, 3H), 2.19-2.06 (m, 5H), 1.97-1.81 (m, 5H), 1.47 (m, 4H), 1.22-1.16 (m, 2H), 0.88 (t, J=7.3 Hz, 3H).
Compound 36 (17 mg, 26 μmol, 1.0 eq) and compound McGGFG (18 mg, 29 μmol, 1.1 eq) were dissolved in DMF (0.5 mL), and HATU (11 mg, 29 μmol, 1.1 eq) and DIEA (10 μL, 58 μmol, 2.2 eq) were added, and stirred at room temperature for 3 hours. The reaction solution was directly purified by reversed-phase column chromatography (40% CH3CN in H2O, 0.05% HCOOH) to obtain a white solid (6 mg, 20%). MS (ESI): m/z found Fragment 1, Positive=541.0 Fragment 2, Positive=591.0.
1H NMR (600 MHz, DMSO-d6) δ 8.60-8.53 (m, 2H), 8.28 (t, J=5.5 Hz, 1H), 8.14 (d, J=7.9 Hz, 1H), 8.06 (t, J=6.8 Hz, 1H), 8.00 (t, J=5.7 Hz, 1H), 7.82 (d, J=10.9 Hz, 1H), 7.33 (s, 1H), 7.24 (hept, J=5.6, 4.9 Hz, 4H), 7.18 (q, J=7.6 Hz, 1H), 7.00 (s, 2H), 6.53 (s, 1H), 5.58 (dt, J=9.0, 4.5 Hz, 1H), 5.46-5.39 (m, 2H), 5.26-5.18 (m, 2H), 4.58 (dd, J=10.3, 6.8 Hz, 1H), 4.51 (dt, J=23.5, 8.2 Hz, 2H), 4.22 (dd, J=8.2, 4.5 Hz, 1H), 4.08 (s, 2H), 3.75 (q, J=4.3, 3.7 Hz, 1H), 3.71 (d, J=5.0 Hz, 1H), 3.69 (d, J=5.6 Hz, 1H), 3.65 (d, J=5.7 Hz, 2H), 3.58 (dd, J=16.7, 5.4 Hz, 1H), 3.50 (d, J=7.6 Hz, 1H), 3.41 (q, J=7.6 Hz, 1H), 3.37 (d, J=7.1 Hz, 2H), 3.17 (q, J=14.3, 11.5 Hz, 2H), 3.07-3.01 (m, 1H), 2.79 (dd, J=13.9, 9.7 Hz, 1H), 2.41 (s, 3H), 2.14-1.99 (m, 6H), 1.88 (ddt, J=17.5, 14.3, 6.6 Hz, 4H), 1.46 (h, J=7.3 Hz, 4H), 1.18 (p, J=7.9 Hz, 2H), 0.88 (q, J=7.0 Hz, 3H).
The anti-human TROP-2 monoclonal antibody was prepared according to the following method: the heavy chain and light chain genes synthesized by Nanjing GenScript Biotechnology Co., Ltd. were constructed into the pCGS3 expression vector, the endotoxin-free plasmid extraction kit (Tiangen Biochemical Technology Co., Ltd., item number: DP 117) was used to extract the plasmid, and the specific operation was carried out according to the instructions. HEK293f cells were cultured using KOP293 cell culture medium (Zhuhai Kairui, item number: K03252), placed in a 37° C., 5% CO2 shaker, with a rotation speed of 100-130 rpm, and a humidity control of more than 75%.
The day before transfection, HEK 293f cells in logarithmic growth phase and in good growth state were passaged to 2×106/mL, cultured overnight on a shaker (110 rpm, 37° C., 5% CO2), and transfected the next day. Before transfection, preheat TA-293 (293 cell suspension chemical transfection reagent) and KPM (serum-free cell transfection buffer solution) at room temperature, and measure the cell density and viability. The density is 4×106/mL, and the viability is greater than 97% can be used. Transfection was performed according to the KOP293 Transient Transfection Protein Expression System User Guide, and the expression supernatant was harvested by centrifugation 5 days after transfection.
The expression supernatant was filtered with a 0.45 μM filter membrane, and an antibody with an Fc domain was obtained from the expression supernatant using an affinity chromatography column. Equilibrium buffer is 9.5 mM sodium dihydrogen phosphate plus 40.5 mM disodium hydrogen phosphate, pH 7.4; elution buffer is 0.1M glycine, pH 3.0. The eluted antibody was replaced with PBS buffer at pH=7.2.
The interchain disulfide bonds were reduced by adding 1 mM EDTA (invitrogen, item number: AM9260G) and 8-fold molar equivalent of TECP (Thermo, item number: 77720) to the 10 mg/mL Trop-2 monoclonal antibody solution, and the mixture was stirred at 37° C. for 2 hours. The mixture was cooled to the target temperature of 4° C. and 10 drug equivalents per mole of compounds 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 were added as a 10% (v/v) solution in DMSO (Sigma, Cat. No.: D2660). After 2 hours, the antibody-drug conjugate was obtained by ultrafiltration with PBS, named TROP-2-ADC-1, TROP-2-ADC-2, TROP-2-ADC-3, TROP-2-ADC-4, TROP-2-ADC-5, TROP-2-ADC-6, TROP-2-ADC-7, TROP-2-ADC-8, TROP-2-ADC-9, TROP-2-ADC-10, TROP-2-ADC-11, TROP-2-ADC-12, TROP-2-ADC-13, TROP-2-ADC-14, TROP-2-ADC-15, TROP-2-ADC-16, TROP-2-ADC-17, TROP-2-ADC-18, TROP-2-ADC-19, TROP-2-ADC-20, TROP-2-ADC-21, TROP-2-ADC-22, TROP-2-ADC-23 in order. After the ultrafiltration was completed, DAR (Drug-to-Antibody Ratio) was detected by using the absorbance method at 280 nm and 365 nm and HIC-HPLC and the results are shown in Table 3.
The anti-human HER3 monoclonal antibody was prepared according to the following method: the heavy chain and light chain genes synthesized by Nanjing GenScript Biotechnology Co., Ltd. were constructed into the pCGS3 expression vector. The plasmid was extracted using an endotoxin-free plasmid extraction kit (Tiangen Biochemical Technology Co., Ltd., catalog number: DP 117), and the specific operation was performed according to the instructions. HEK293f cells were cultured using KOP293 cell culture medium (Zhuhai Kairui, product number: K03252), placed in a 37° C., 5% CO2 shaker, with a rotation speed of 100-130 rpm, and a humidity control of more than 75%. The day before transfection, HEK 293f cells in logarithmic growth phase and in good growth state were passaged to 2×106/mL, cultured overnight on a shaker (110 rpm, 37° C., 5% CO2), and transfected the next day. Before transfection, preheat TA-293 (293 cell suspension chemical transfection reagent) and KPM (serum-free cell transfection buffer solution) at room temperature, and measure the cell density and viability. The density is 4×106/mL, and the viability is greater than 98% can be used. Transfection was performed according to the KOP293 Transient Transfection Protein Expression System User Guide, and the expression supernatant was harvested by centrifugation 6 days after transfection.
The expression supernatant was filtered with a 0.45 μM filter membrane, and an antibody with an Fc domain was obtained from the expression supernatant using an affinity chromatography column. Equilibrium buffer is 9.5 mM sodium dihydrogen phosphate plus 40.5 mM disodium hydrogen phosphate, pH 7.4; elution buffer is 0.1M glycine, pH 3.0. The eluted antibody was replaced with PBS buffer at pH=7.2.
By adding 1 mM EDTA (invitrogen, item number: AM9260G) and 8-fold molar equivalent of TECP (Thermo, item number: 77720) to the 10 mg/mL HER3 monoclonal antibody solution to reduce the interchain disulfide bonds, the mixture was stirred at 37° C. and 250 rpm for 2.5 hours. The mixture was cooled to the target temperature of 4° C. and 10 drug equivalents per mole of compounds 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 72, 73, 74, and 75 were added as a 1000 (v/v) solution in DMSO (Sigma, Cat. No.: D2660). After 3 hours, replace antibody-drug conjugates with PBS ultrafiltration, named HER3-ADC-1, HER3-ADC-2, HER3-ADC-3, THR3-ADC-4, THR3-ADC-5, HLER3-ADC-6, THR3-ADC-7, THR3-ADC-8, HER3-ADC-9, HER3-ADC-10, HER3-ADC-11, ER3-ADC-12, ER3-ADC-13, ER3-ADC-14, THR3-ADC-15, HER3-ADC-16, THR3-ADC-17, THR3-ADC-18, HER3-ADC-19, HER3-ADC-20, TER3-ADC-21, TER3-ADC-22, TER3-ADC-23, HER3-ADC-24, HER3-ADC-25, HER3-ADC-26, HER3-ADC-27. The specific structure of the antibody conjugates are shown in Table 4.
Table 4 structural formulas, of the HRR3-ADC,
The preparation method of the positive reference drug U3-1402 (PATRITUMAB DERUXTECAN) used in the test example of the present invention is prepared by referring to the preparation method of the antibody-drug conjugate (16a) in patent CN106163559B.
The cell line used in this experiment is the MDA-MB-468 cell line with high TROP-2 expression (Nanjing Cobioer Biosciences Co., Ltd., catalog number: CBP60387). Prepare cell suspension with fresh cell culture medium containing 10% FBS at a density of 3×103 cells/mL, add 90 μL per well into an all-white 96-well cell culture plate (Beyotime, item number: FCP968-80pcs), 5% carbon dioxide 37° C. for 24 h. Compounds 1-36 were formulated with DMSO to 1 mM, respectively. Add 20 μL of different samples to be tested in the first column of the 96-well U-bottom dispensing plate, and the sample concentration is 1000 uM; add 20 μL DMSO to each well of the second to tenth columns. Take 5 μL of the sample from the first column to 20 μL DMSO in the second column, mix well, take 5 μL to the third column, and so on to the ninth column. Take 5 μL of the medicine from each well of the dispensing plate to 95 μL of DMEM medium, and mix well. Add 10 μL of prepared samples to be tested at different concentrations to the culture plate, and duplicate wells for each sample. 5% carbon dioxide and cultured at 37° C. for 3 days. Add 50 μL of CTG (CellTiter-Glo® Luminescent Cell Viability Assay, Promega, item number: G7573) to each well, mix on a shaker at room temperature for 30 minutes, incubate for 10 minutes, and read chemiluminescence on a microplate reader (TECAN, Spark). Graphpad Prism 8 software used for data analysis. The experimental results are shown in Table 6.
Conclusion: the Exatecan derivatives provided by the present invention have obvious proliferation inhibitory activity on MDA-MB-468 cells.
The cell line used in this test example was purchased from Nanjing Cobioer Biosciences Co., Ltd. MDA-MB-468 cells, item number: CBP60387, TROP-2 high expression cell line.
The cell suspension was prepared with FACS (2% FBS+PBS) staining buffer, and the density was 2×106 cells/mL, the cell suspension was spread on a 96-well U-bottom cell plate (UWP043096) at 50 μL/well, and each sample was set up a set of parallel experiments. Dilute TROP-2 antibody (naked antibody) or TROP-2-ADC compound (TROP2-ADC-3, TROP2-ADC-4, TROP2-ADC-19, TROP2-ADC-21 or TROP2-ADC-23) to a concentration of 20 ug/ml. Take 50 μl of TROP-2 antibody or TROP-2-ADC compound dilutions and add them to 96-well U-bottom cell plates, mix gently and then ice-bath for 60 minutes. Add 200 μL of FACS staining buffer to wash the cells, centrifuge at 300 g at 4° C. for 5 mins, discard the supernatant, and repeat the washing twice. Add 30 μL of FACS to each well to resuspend the cells, and incubate at 37° C. for 15 min, 30 min, 60 min, and 90 min to internalize the antibodies bound to the cell surface. At each time point, another sample incubated for the same time at 4° C. was used as a negative control for no internalization. At each time point, transfer the corresponding 37° C. and 4° C. incubated samples to ice and add 170 μL of ice FACS to terminate internalization. Cells were precipitated by centrifugation at 300 g for 5 min at 4° C. Add secondary antibody at a ratio of 0.5:100, 100 μL/well. Incubate at 4° C. for 30 min in the dark. Add 200 μL FACS staining buffer to wash the cells and remove unbound antibody. Centrifuge at 300 g at 4° C. for 5 min to precipitate the cells, and discard the supernatant. Repeat the wash 2 times. Resuspend the stained cells in 100 μL ice-cold PBS. After washing the cells, they were analyzed by flow cytometry.
The reduction in mean fluorescence intensity (MFI) of samples incubated at 37° C. relative to samples incubated at 4° C. can be obtained by the following formula:
According to the calculated percentage of internalization, a line graph of the percentage of internalization was drawn, as shown in
In this test example, the affinity of the TROP-2 antibody and the TROP-2-ADC compound obtained according to the above examples to TROP-2 is detected by means of capturing antibody.
Use the Sensor Chip Protein A biosensor chip (Cytiva) to capture the antibody (TROP-2 antibody or TROP-2-ADC compound), then flow through the antigen TROP-2-his on the surface of the chip, and use the Biacore K8 instrument (Cytiva) Real-time detection of reaction signals to obtain association and dissociation curves. After the dissociation of each experiment cycle, the chip was washed and regenerated with regeneration buffer. After the experiment, GE Biacore K8 Evaluation version 3.0 software was used to fit the data with the (1:1) Langmuir model to obtain the affinity value. The affinity between antibody and protein is shown in Table 7:
As shown in Table 7, the KD values of the naked antibody and the TROP-2-AD compounds of the present invention are both in the picomolar range, and there is no significant difference between the two.
The cell line used in this experiment is the MDA-MB-468 cell line with high TROP-2 expression (Nanjing Cobioer Biosciences Co., Ltd., catalog number: CBP60387). Prepare cell suspension with fresh cell culture medium containing 1000 FBS at a density of 2×105 cells/mL, add 100 μL per well into an all-white 96-well cell culture plate (Beyotime, product number: FCP968-80pcs), and culture with 5% carbon dioxide at 37° C. for 24 h. ADC samples were made up to 10 μM in PBS. Take this as the first concentration, and use PBS for five-fold serial dilution, a total of 9 concentrations. Aspirate 50 μL of culture solution from each well, and then add 50 μL of the above-mentioned ADC solution, so the ADC concentration in the first well is 5M, and the final volume is 100 μL per well. 5% carbon dioxide and cultured at 37° C. for 3 days. Add 100 μL of CTG (CellTiter-Glo® Luminescent Cell Viability Assay, Promega, item number: G7573) to each well, mix on a decolorizing shaker at room temperature for 30 min, incubate for 10 minutes, read chemiluminescence on a microplate reader (TECAN, Spark), and use Graphpad Prism 5 software for data analysis. The experimental results are shown in Table 8.
Conclusion: the antibody-drug conjugates against TROP-2 target of the present invention has obvious proliferation inhibitory activity on TROP-2 positive cell MDA-MB-468 cells.
Human triple-negative breast cancer cell MDA-MB-468 (Nanjing Cobioer Biosciences Co., Ltd., item number: CBP60387) was inoculated subcutaneously in the right flank of BALB/c-Nude nude mice (5×106/200 μL/mouse, with 50% low growth factor artificial basement membrane). After the cells were inoculated, the tumor grew for 10 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (DO), 5 animals in each group, 12 groups in total.
Tail vein injection was used, the control group was given PBS, and the experimental group was given the TROP-2-ADC compounds TROP2-ADC-3, TROP2-ADC-19, TROP2-ADC-21 or TROP2-ADC-23 provided by the present invention. The multiple doses are 5 mg/kg, 2 mg/kg, 1 mg/kg, twice a week, for 2 weeks. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (PBS) and the experimental group at the end of the experiment, respectively. The results are shown in
Human orthotopic pancreatic cancer cells Bxpc-3 (Cobioer) (4×106/200 μL/mouse, with 50% low growth factor artificial basement membrane) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the cells were inoculated, the tumor grew for 6 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (Do), 5 animals in each group, 4 groups in total.
Adopt tail vein injection administration, the control group applies PBS, the experimental group applies the TROP-2-ADC-21 compound provided by the present invention, and the administration dosage is 3 mg/kg, 6 mg/kg, 12 mg/kg, 2 times a week, 5 times. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (PBS) and the experimental group at the end of the experiment, respectively. The results are shown in
Human gastric cancer cells NCI-N87 (Cobioer) (5×106/200 μL/mouse, with 50% low growth factor artificial basement membrane) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the cells were inoculated, the tumor grew for 3 days, and when the tumor volume grew to about 130 mm3, the animals were randomly divided into groups (Do), 5 animals in each group, 4 groups in total.
Adopt tail vein injection administration, the control group applies PBS, and the experimental group applies the TROP-2-ADC-21 compound provided by the present invention. The administration dosage is 3 mg/kg, 6 mg/kg, 12 mg/kg, 2 times a week for 2 weeks. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (PBS) and the experimental group at the end of the experiment, respectively. The results are shown in
The cell lines used in this experiment were human breast cancer cell line HCC1569, human colorectal adenocarcinoma cell line SW620, human malignant melanoma cell line A375, and human non-small cell lung cancer cell line NCI-H358 with high expression of HER3. The above cell lines were inoculated into a 96-well plate at a density of 103 cells/100 μL/well, and 200 μL PBS was added to the outermost circle to reduce the volatilization of the medium. Incubate at 37° C., 5% CO2 for 48 h and remove the 96-well plate. 100 μL of diluted ADC samples were added to the wells, and the ADC samples were prepared to 10 μM with PBS, which was used as the first concentration, and five-fold gradient dilutions were made with PBS, a total of 9 gradients, and the concentration at the last point was 0. Incubate at 37° C., 5% CO2 for 120 h. Take out the 96-well plate, remove the culture solution, and add 100 μL of detection solution (CCK8:culture solution=1:9) to each well; place it in a 5% CO2 incubator at 37° C. for 4 hours; and use a microplate reader (TECAN, Cat. No.: CEY0017) to read the value at 450 nm, record it with Excel, and use Graphpad Prism 5 software for data analysis. The experimental results are shown in Table 9-Table 12.
Conclusion: The antibody-drug conjugates against HER3 target of the present invention have obvious proliferation inhibitory activity on HER3 positive cells SW620 cells, HCC1569 cells, A375 cells and NCI-H358 cells.
Human breast cancer cells MDA-MB-453 (Cobioer) (5×106/200 μL/mouse, with 50% low growth factor artificial basement membrane) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the cells were inoculated, the tumor grew for 10 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (denoted as day 0, ie., DO), with 5 animals in each group, 10 groups in total. It is administered by tail vein injection, with multiple doses of 5 mg/kg and 10 mg/kg administered twice a week for 2 weeks. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (PBS) and the experimental group at the end of the experiment, respectively. The tumor growth curve is shown in
Human colorectal adenocarcinoma cells SW620 (5×106/200 μL/mouse) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the tumor grew for 8 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (denoted as day 0, ie., D0), with 5 animals in each group, 14 groups in total. Tail vein injection was used, and multiple doses of 3 mg/kg, 6 mg/kg and 12 mg/kg were administered, twice a week for 2 weeks. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (PBS or vehicle) and the experimental group at the end of the experiment, respectively. The inhibitory effect of the HER3-ADC compound provided by the present invention on tumors in animals shows a positive correlation with the dose, and the tumor growth curve is shown in
% MFI at tx time point=MFI of samples incubated at 37° C.×100/MFI of samples incubated at 4° C.;
Percentage of internalization at tx time point=100−% MFI at tx time point
Draw a line graph of the percentage of internalization based on the percentage of internalization calculated. The endocytosis of the HER3-ADC compound provided by the present invention is the key to the efficacy of the ADC. In this experiment, flow cytometry was used to detect the internalization level of the naked antibody and the HER3-ADC compound in cells HCC1569, SW620, and MDA-MB-453. The results are shown in
Taking the HER3 monoclonal antibody (naked antibody) and the ADC compounds after coupling the drug provided by the present invention as the detection target
The binding ability of naked antibody, HER3-ADC-19, HER3-ADC-20, HER3-ADC-21 to human HER3-His antigen was determined at 7 different concentrations (0.625-40 nM) using protein A biosensor chip. As shown in Table 13, the KD values of both naked antibody and HER3-ADC compounds were in the nanomolar range, and there was no significant difference between them.
Melanoma cell A375 (derived from ATCC, 5×106/200 μL/mouse, with 50% low growth factor artificial basement membrane) was inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the cells were inoculated, the tumor grew for 8 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (denoted as DO, that is, day 0), with 5 animals in each group, 4 groups in total. Tail vein injection was used, and the tested drugs were HER3-ADC-21 (5 mg/kg), positive control drug U3-1402 (5 mg/kg), HER3 monoclonal antibody (10 mg/kg) and blank control group (PBS), 2 times a week for 2 weeks. Tumor volume was measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (Vehicle) and the experimental group at the end of the experiment, respectively. The results were shown in
According to the results in
Human non-small cell lung cancer cells NCI-H358 (derived from ATCC, 3×106/200 μL/mouse, with 50% low growth factor artificial basement membrane) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. After the cells were inoculated, the tumor grew for 5 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (denoted as DO, ie., day 0), with 5 animals in each group, 4 groups in total. Tail vein injection was used, and the tested drugs were HER3-ADC-21 (10 mg/kg), positive control drug U3-1402 (10 mg/kg), HER3 monoclonal antibody (10 mg/kg) and blank control group (PBS), 2 times a week for 2 weeks. Tumor volume was measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (Vehicle) and the experimental group at the end of the experiment, respectively. The result is shown in
The results shown in
According to the results shown in
According to the experimental results shown in
In this experiment, HCC1569 cells (HER3 positive cell line) and Nalm6-GFP (HER3 negative cell line) cells were co-cultured to detect the bystander killing ability of HER3-ADC-21.
Calculation formula: Killing rate=(cell viability rate without drug application*cell positive rate−cell viability rate with drug application*cell positive rate)/cell viability rate without drug application*cell positive rate, wherein the cell positive rate refers to the percentage of Nalm6 cells Ratio in co-cultured cells (HCC1569 cells and Nalm6-GFP cells). The results are shown in Table 17.
According to the results shown in Table 17, when cultured alone, HER3-ADC-21 has no killing effect on Nalm6-GFP (HER3-negative cell line) cells, but has a killing effect on HCC1569 (HER3-positive cell line) cells. After HCC1569 (HER3-positive cell line) cells and Nalm6-GFP (HER3-negative cell line) cells were co-cultured, HER3-ADC-21 had a killing effect on Nalm6-GFP (HER3-negative cell line) cells. In summary, HER3-ADC-21 has a bystander effect.
MDA-MB-468 (human breast cancer cells, Nanjing Cobioer, CBP60387, TROP-2 positive cells) and Nalm6-GFP cells (human B lymphoid leukemia cells, purchased from Creative Biogene, CSC-RR0360, TROP-2 negative cells) were culture using DMEM/low glucose+10% FBS and RPMI1640+10% FBS respectively, trypsinize the cells, neutralize with fresh medium, centrifuge at 300 g for 5 minutes, discard the supernatant, and resuspend the cells in RPMI1640+10% FBS. After cell counting, 200,000 MDA-MB-468 cells/well and 100,000 Nalm6-GFP cells/well were spread in a 6-well plate according to the ratio of MDA-MB-468:Nalm6-GFP=2:1. In addition, two groups of cells containing only MDA-MB-468 and Nalm6-GFP were used for control experiments. After 24 hours of incubation, ADC compounds were added at drug concentrations of 5 nM, 10 nM, 20 nM, 40 nM, 80 nM. After continuing to incubate for 5 days, detection was performed. For detection, cells were collected and washed three times with PBS. 100 μL NIR (Corning Incorporated Costor, 3590) staining solution (diluted 1:1000) was stained in the dark for 15 min, washed once with PBS, and flow cytometric detection was performed.
According to the streaming results, calculate the killing rate: Killing rate=(cell viability rate without drug application*cell positive rate-cell viability rate with drug application*cell positive rate)/cell viability rate without drug application*cell positive rate; the cell positive rate refers to the percentage of Nalm6-GFP cells in the total ratio of cultured cells (MDA-MB-468 cells and Nalm6-GFP cells). The results are shown in Table 18 and Table 19.
According to the results in Table 18, the TROP-2-ADC molecule of the present invention has obvious killing effect on TROP-2 positive cells, but has no obvious inhibitory effect on TROP-2 negative cells. In cells co-cultured with TROP-2 positive cells and TROP-2 negative cells, the ADC molecule of the present application can significantly inhibit both TROP-2 positive and TROP-2 negative cells, showing obvious bystander killing effect. TROP-2-ADC drugs also have a bystander effect. The results in Table 19 show that other TROP-2-ADC molecules of the present invention also have obvious bystander killing effects.
Human colorectal adenocarcinoma cells SW620 (5×106/200 μL/mouse) were inoculated subcutaneously in the right flank of BALB/c-Nude nude mice. The tumor grew for 8 days, the tumor volume grew to about 130 mm3, and the animals were randomly divided into groups (denoted as DO, ie., day 0), with 5 animals in each group, 11 groups in total. It is administered by tail vein injection, with a single dose of 10 mg/kg, twice a week, for 2 weeks. Tumor volume and body weight were measured twice a week, and the data were recorded. Excel 2016 statistical software is used for data statistics: average value is calculated by average; SD value is calculated by STDEV; SEM value is calculated by STDEV/SQRT. Tumor growth curves were made using GraphPad Prism 8.0.2.2.263 software.
Wherein, V0 and VT are the tumor volumes at the beginning of the experiment and at the end of the experiment, respectively. CRTV and TRTV are the relative tumor volumes of the blank control group (Vehicle) and the experimental group at the end of the experiment, respectively. The results are shown in
The present invention has been described through the above-mentioned examples, but it should be understood that the above-mentioned examples are only for the purpose of illustration and description, and are not intended to limit the present invention to the scope of the described embodiments. In addition, those skilled in the art can understand that the present invention is not limited to the above-mentioned examples, and more variations and modifications can be made according to the teachings of the present invention, and these variations and modifications all fall within the claimed scope of the present invention. within the range. The protection scope of the present invention is defined by the appended claims and their equivalent scope.
| Number | Date | Country | Kind |
|---|---|---|---|
| 202210062910.7 | Jan 2022 | CN | national |
| 202211318840.3 | Oct 2022 | CN | national |
| Filing Document | Filing Date | Country | Kind |
|---|---|---|---|
| PCT/CN2023/073014 | 1/18/2023 | WO |