CN115003684A - Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same - Google Patents

Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same Download PDF

Info

Publication number
CN115003684A
CN115003684A CN202080094623.3A CN202080094623A CN115003684A CN 115003684 A CN115003684 A CN 115003684A CN 202080094623 A CN202080094623 A CN 202080094623A CN 115003684 A CN115003684 A CN 115003684A
Authority
CN
China
Prior art keywords
binding peptide
compound
peptide conjugate
isotopically labeled
ipr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202080094623.3A
Other languages
Chinese (zh)
Inventor
张君革
晏良增
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mainstream Biotechnology Shanghai Co ltd
Original Assignee
Mainline Biosciences Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/752,690 external-priority patent/US11123437B2/en
Application filed by Mainline Biosciences Ltd filed Critical Mainline Biosciences Ltd
Publication of CN115003684A publication Critical patent/CN115003684A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/008Peptides; Proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids

Abstract

The present invention provides a selective CXCR4 binding peptide conjugate ("PC") and methods of use and preparation thereof. In particular, the selective CXCR4 binding peptide conjugates of the invention include a peptide moiety that selectively binds CXCR4 and a compound of pharmaceutical value (e.g., an imaging agent, a diagnostic agent, or a therapeutically or pharmaceutically active compound). In one particular embodiment, the selective CXCR4 binding peptide conjugate ("PC") is of the following structural formula: (SEQ ID NO:1) or a pharmaceutically acceptable salt thereof, wherein a, b, AA 1 、AA 2 、Ar 1 、X 1 And AA 3 As defined herein. The peptide conjugates of the present invention are useful for a variety of medical uses, including but not limited to targeted delivery or imaging of patients, or diagnosing diseases or clinical conditions associated with overexpression and/or upregulation of CXCR4, such as cancer, HIV infection, and immune diseases. Also disclosed herein are compositions, kits and methods for such uses.

Description

Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same
Technical Field
The present invention relates to isotopically labeled selective CXCR4 binding peptide conjugates ("PCs") and methods of use and production thereof. In particular, isotopically-labeled selective CXCR4 conjugate peptide conjugates of the present invention are provided using positron-emitting radioisotopes, radioactive metal isotopes, deuterium, tritium, and, 13 C、 14 C、 18 O or a combination thereof, and comprises a peptide moiety that selectively binds CXCR4 and a compound of pharmaceutical value, such as an imaging agent, a diagnostic agent, or a therapeutically active or pharmaceutically active compound.
Background
CXCL12 (also known as stromal cell derived factor-1 or SDF-1) and CXCR4 (a chemokine and chemokine receptor pair) play important roles in the various stages of hematopoiesis, tumorigenesis, and embryonic development. For example, activation of CXCR4 by CXCL12 suggests that chemotaxis of leukocytes in the immune system can be directed during embryonic development to address inflammation and progenitor cell migration. Activation of CXCR4 by CXCL12 has also been shown to mediate signaling pathways involved in breast cancer metastasis and memory T cell migration.
CXCR4 is a G protein-coupled receptor, also known as fusion or CD184 (cluster of differentiation 184), constitutively expressed or overexpressed in a variety of human cancers, promoting local tumor cell proliferation, survival and angiogenesis. CXCR4 is reported to be a co-receptor for HIV entry into and infection of host cells and has been evaluated as a potential treatment for HIV.
Reports confirm that CXCR4 is overexpressed in many human cancers. CXCR4 antagonism has been shown to disrupt tumor-stromal interactions, sensitize cancer cells to cytotoxic drugs, and reduce tumor growth and metastatic burden. Thus, CXCR4 is not only a target for potential therapeutic intervention in cancer therapy, but is also a target for noninvasive monitoring of disease progression, therapeutic guidance, and other diagnostic purposes. Some even believe that binding to and interaction with CXCR4 is a potential means of targeted delivery.
Thus, compounds having a moiety that selectively binds CXCR4 (i.e., CXCR4 selective binding conjugates) are believed to have a wide range of uses, including but not limited to the treatment of various clinical conditions, diagnosis of patients, and medical imaging associated with activation or overexpression of CXCR 4. Furthermore, by labeling moieties that can selectively bind CXCR4, various modes of drug delivery, drug interactions, diagnostics, in vivo imaging of cells affected by overexpression of CXCR4, and the like can be studied. It would be further useful if such a marker compound did not significantly alter the interaction with CXCR 4.
Thus, there is a need for CXCR4 conjugates that are capable of selectively binding to compounds of pharmaceutical value. Furthermore, there is a need for a labeled conjugate that does not significantly change physical properties (e.g., binding constant, drug activity, its three-dimensional structure, etc.).
Disclosure of Invention
The present invention provides isotopically-labeled selective CXCR4 binding peptide conjugates ("PCs") and methods of use and production thereof. In particular, the selective CXCR4 binding peptide conjugates of the invention are useful as positron emitting radioisotopes, radiometal isotopes, deuterium, tritium, and, 13 C、 14 C、 18 O and/or combinations thereof, and comprises a peptide moiety that selectively binds CXCR4 and a compound of pharmaceutical value, such as an imaging agent, a diagnostic agent, or a therapeutically active or pharmaceutically active compound. In some embodiments, the peptidyl moiety of the compounds of the invention is substituted with a positron-emitting radioisotope, a radiometal isotope, deuterium, tritium, a, 13 C、 14 C、 18 O or a combination thereof. In this way, one can easily track, image or study compounds in vivo. The peptide or peptidyl moiety is linked to a compound of pharmaceutical value through a polymer linker. The peptide conjugates of the invention are useful for a variety of medical uses, including but not limited to targeted drug delivery, patient imaging, in vivo studies of compounds of medical value (e.g., with C)XCR4 interactions, etc.), diagnosing a disease or clinical condition in a patient associated with overexpression and/or upregulation of CXCR4, such as cancer, HIV infection, and immune diseases. Also disclosed herein are compositions, kits and methods for such uses.
In a particular aspect of the invention, isotopically labeled selective CXCR4 binding peptide conjugates ("PCs") of the following structural formula are provided:
Figure BDA0003765213480000021
or a pharmaceutically acceptable salt thereof,
wherein:
a is 0 or 1;
b is an integer between 1 and 4;
AA 1 and the sulfur atom to which it is attached is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine;
AA 2 and the sulfur atom to which it is attached is cysteine or homocysteine;
Ar 1 is an optionally substituted aryl group;
X 1 is Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or Lys (iPr);
AA 3 each is independently Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), Lys (iPr);
and wherein AA 1 、X 1 、AA 2 Or AA 3 Comprises a moiety of formula (la):
–L 1 –Q,
wherein
L 1 Is a polymer linker having a functional group for linking Q, wherein L 1 Comprising from about 2 to about 20 monomers or copolymers;
q is absent, is AA 4 Or is of the formula- [ AA 4 -Y 1 -L 2 ] c -Y 2 A part of-Z and a part of-Z,
wherein
c is 0 or 1;
AA 4 is an amino acid or a derivative thereof;
Y 1 is amino acid AA 4 A side chain functional group of (1);
L 2 is a non-polymeric linker;
z is a compound having a pharmaceutical value; and is
Y 2 Is a functional group or L of the compound having medical value 2 A functional group of (a);
wherein, AA 1 、AA 2 、X 1 、AA 3 And AA 4 With one or more of a positron-emitting radioisotope, a radioactive metal isotope, deuterium, tritium, a, 13 C、 14 C、 18 O or a combination thereof.
Compounds of pharmaceutical value useful in the present invention include, but are not limited to, compounds useful in the diagnosis, treatment or analysis of various clinical conditions. Exemplary compounds of medical value include, but are not limited to, imaging agents, contrast agents, and therapeutic agents (e.g., drugs).
In some embodiments, the selective CXCR4 binding peptide conjugates of the invention have the following structural formula:
Figure BDA0003765213480000041
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、L 1 Q and Ar 1 As defined herein. In the present invention, when referring to variables, the terms "defined herein" and "defined above" incorporate by reference the broad definition of the variables as well as any narrow definition, including preferred, more preferred and most preferred definitions (if any).
In some embodiments, Q is absent from a compound of formulae disclosed herein. In this case, AA 1 、X 1 、AA 2 Or AA 3 At least one of which comprises the structural formula L 1 Part (c) of (a). Such compounds are useful as linking AA 4 、–AA 4 –Y 1 –L 2 Or of the formula [ AA ] 4 –Y 1 –L 2 ] c –Y 2 -a starting material for a part of Z.
In other embodiments, Q is AA 4 . Such compounds may be used in the formula-Y of the link (attachment) or the link (link) 1 -L 2 -Y 2 -a moiety of Z.
In yet other embodiments, Q is structural formula (xxxvi): - [ AA 4 –Y 1 –L 2 ] c –Y 2 -a portion of Z. Accordingly, the present invention provides starting materials for linking compounds of pharmaceutical value as well as selective CXCR4 binding peptide conjugates comprising at least one compound of pharmaceutical value.
In other embodiments, a is 0. In other embodiments, a is 1.
In still other embodiments, X 1 And AA 3 Is isotopically labelled. In other embodiments, X 1 And AA 3 Are all isotopically labelled. Exemplary isotopic labels include, but are not limited to, deuterium, tritium, 13 C、 14 C、 18 O and combinations thereof. In a particular embodiment, the isotopic label comprises deuterium or tritium. Typically, the isotope is labeled X 1 And/or AA 3 Containing deuterium, tritium, 13 C、 14 C、 18 O or a combination thereof. In a particular embodiment, X 1 And AA 3 Is isotopically labelled with deuterium or tritium. In one embodiment, X 1 And AA 3 Is isotopically labeled lysine (iPr). More specifically, in one embodiment, X 1 And AA 3 Are all lysine (deuterated iPr).
In still other embodiments, the compound of pharmaceutical value is a therapeutic agent. In some embodiments, the therapeutic agent is an anti-cancer agent. Exemplary anticancer agents that may be used in the present invention include, but are not limited to, paclitaxel, altretamine, asparaginase, bleomycin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, diethylstilbestrol, eribulin, ethinylestradiol, etoposide, mitomycin, o-chlorobenzenedichloroethane, mitoxantrone, pentostatin (Pentastatin), bromopropiperazine, mithramycin, prednisone, procarbazine, streptozotocin, tamoxifen, teniposide, vinblastine, and vincristine. In general, any anti-cancer agent having a functional group suitable for attachment may be used. Such anti-cancer agents will be apparent to those skilled in the art upon reading the present invention.
In other embodiments, isotopically labeled selective CXCR4 binding peptide conjugates of the invention have the structure:
Figure BDA0003765213480000051
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、AA 4 、Y 1 、Y 2 、L 2 Z and Ar 1 Are those defined herein; l is 1a Is a polymer linker comprising from about 2 to about 20 monomers or copolymers.
In some embodiments, L is 2 Is a non-polymeric linker of the formula:
–CH 2 –C(=O)–NH–CH 2 –、–CH 2 –NH–C(=O)–CH 2 –、–(CH 2 ) n –C(=O)–NH–CH 2 –、–(CH 2 ) n –NH–C(=O)–CH 2 -, or- (CH) 2 ) n -, where n is an integer between 1 and 6.
In still other embodiments, AA 1 Together with the sulfur atom attached thereto is an optionally substituted cysteine or an optionally substituted homocysteine. For example, AA 1 The amino group of (A) may be substituted with an acyl group (e.g., acetyl, etc.), an alkyl group (e.g., acetyl, etc.), or a salt thereofMethyl, dimethyl, ethyl, dimethyl, propyl, dipropyl, isopropyl, di (isopropyl), etc.), haloalkyl (e.g., trifluoromethyl, difluoromethyl, etc.). For those skilled in the art who have read this disclosure, AA 1 Other suitable substituents will be apparent.
And AA 1 Similarly, AA 2 And the sulfur atom to which it is attached may also be an optionally substituted cysteine or an optionally substituted homocysteine.
In a particular embodiment, X 1 And/or AA 3 Is Lys (iPr) or Lys (deuterated iPr). When deuterated (e.g., "deuterated iPr"), isopropyl ("iPr") can have one or more hydrogen atoms replaced with deuterium. In some embodiments, at least 2, typically at least 3, typically at least 4, more typically at least 5, and most typically at least 6 hydrogen atoms of the iPr are replaced with deuterium atoms. In the present invention, the abbreviation of amino acid AA (X) refers to the amino acid AA substituted with "X" on the side chain functional group. For example, lysine (iPr) refers to lysine substituted with isopropyl on the amine functionality of the lysine side chain.
In other embodiments of the present invention, Ar 1 Is phenyl. In a particular embodiment, Ar 1 The structural formula of (A) is:
Figure BDA0003765213480000061
in another particular embodiment of the invention, b is 1. In this embodiment, in some instances, b is 1, AA 3 Is Lys (iPr) or Lys (deuterated iPr).
In another embodiment of the invention, L 1 Is a polymeric linker of the formula:
–NH–(CH 2 ) 2 –[O–CH 2 –CH 2 –] n –O–(CH 2 ) 2 -C (═ O) -, where n is an integer from 2 to 20, typically from 2 to 15, typically from 2 to 10, more typically from 2 to 8, but more typically from 2 to 6, most typicallyIs 3 to 6. In some cases, -NH-linked amino acid AA 3 The carbonyl carbon of (a). In other cases, -C (═ O) -linking amino acid AA 4 The alpha-amino functional group of (a).
In a particular embodiment, AA 4 Is cysteine, homocysteine or penicillin amine.
In other embodiments, AA 1 、X 1 、AA 2 Or AA 3 At least one of the amino acid residues is the (D) -isomer.
In other embodiments, a is 0.
In other embodiments, AA 1 Is homocysteine.
Exemplary selective CXCR4 binding peptide conjugates of the invention include, but are not limited to, compounds of structural formula a, structural formula B, structural formula C, structural formula D, structural formula E, structural formula F, structural formula G, structural formula H, structural formula I, structural formula J, structural formula K, structural formula L, structural formula M, structural formula N, structural formula O, and structural formula P, as well as the corresponding deuterium labeled compounds (i.e., -D12) and tritium labeled compounds (i.e., -T12).
Another aspect of the invention provides a method of treating a subject having cancer, the method comprising administering to the subject a therapeutically effective amount of a selective CXCR4 binding peptide conjugate disclosed herein, wherein the compound of pharmaceutical value is an anti-cancer agent. Exemplary anticancer agents for use in the methods of the invention include, but are not limited to, paclitaxel, altretamine, asparaginase, bleomycin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, diethylstilbestrol, eribulin, ethinylestradiol, etoposide, mitomycin, o-chlorobenzenedichloroethane, mitoxantrone, pentostatin, bromopropylpiperazine, mithramycin, prednisone, procarbazine, streptozotocin, tamoxifen, teniposide, vinblastine, and vincristine.
In some embodiments, the methods of the invention provide for the treatment of a cancer selected from the group consisting of breast cancer, lung cancer, pancreatic cancer, renal cancer, brain cancer, blood cancer, leukemia, prostate cancer, ovarian cancer, and bladder cancer.
Detailed Description
CXCR4 plays an important role in the immune and inflammatory responses of various diseases and conditions, including cancer, viral infections, and autoimmune diseases such as rheumatoid arthritis. The present invention is based, at least in part, on reducing or preventing overexpression or activation of CXCR4 for the treatment, diagnosis, or imaging of clinical conditions associated with overexpression and/or activation of CXCR 4. As used herein, the term "overexpression and/or activation" refers to gene expression above its normal (i.e., control) or baseline level and/or activation of CXCR4 above its normal, control or baseline level, respectively.
The terms "normal", "baseline level" and "control level" are used interchangeably herein and refer to the level of expression and/or activity of CXCR4 in a subject (such as a subject disclosed herein) who is free of a disease or clinical condition associated with overexpression and/or activation of CXCR 4. In some embodiments, the baseline level can be a normal level, meaning a level in a sample from a normal subject who does not have a clinical condition associated with overexpression and/or activation (or activity) of CXCR 4. This allows for the determination based on a baseline level of CXCR4 expression or its biological activity, i.e., whether a sample to be tested or assessed for a disease or clinical condition has measurable increased, decreased or essentially no change in CXCR4 expression or activation as compared to the baseline level.
It is understood that overexpression and/or activation of CXCR4 can also be determined by comparing sample results to positive controls. The term "positive control" as used herein refers to a level of CXCR4 expression and/or activation (or activity) established in a sample based on data from a subject or from a population of individuals who have a disease or clinical condition associated with CXCR4 overexpression and/or activation (e.g., cancer, autoimmune diseases such as rheumatoid arthritis, and viral infections such as HIV infection), where the sample is authentic.
In other embodiments, a baseline level may be established from a previous sample of the subject being tested, so that the subject's progression or regression of the disease may be monitored over time and/or the efficacy of the treatment assessed.
Some aspects of the invention provide compounds having high affinity for CXCR4, which compounds are linked to a diagnostic, therapeutic or imaging agent through a linker. Such compounds include a CXCR4 binding moiety and a pharmaceutically active ingredient (or simply "active compound"). As used herein, the term "pharmaceutically active compound" refers to a compound that has therapeutic activity, or can be used for diagnostic or imaging, or any other use associated with other uses in therapeutic, diagnostic, imaging, analytical, or clinical applications. The invention also provides methods of using the same, e.g., targeted delivery of therapeutic agents to treat clinical conditions caused by or associated with overexpression and/or activation of CXCR4, imaging cells associated with CRCR4 (e.g., in vivo or in vitro), recognizing overexpressed cells or cells that activate CXCR4, and the like, as used herein, the term "high affinity" or "selective" refers to compounds or moieties that bind to CXCR4 having a binding constant (Kb) of about 10nM or less, typically about 3nM or less, and typically 1nM or less. Alternatively, the term "high affinity" or "selective" refers to a compound or moiety that binds to CXCR4 having a half-maximal binding Inhibitory Concentration (IC) of about 30nM or less 50 ) Typically about 10nM or less, and typically about 3nM or less. Determination of binding constants and IC is well known to those skilled in the art 50 The method of (1). See, for example, commonly assigned U.S. provisional patent application No.62/384132 filed on 6/9/2016 and 62/505064 filed on 11/5/2017, and commonly assigned PCT patent application No. PCT/US17/50106 filed on 5/9/2017, all of which are incorporated herein by reference in their entirety. In particular, CXCR4 @, as described in the above provisional patent application, is used 125 I-SDF-1 alpha binding assay to determine K b And IC 50 The value is obtained.
The terms "about" and "approximately" are used interchangeably herein when referring to numerical values and are meant to be within an acceptable error range for the particular value as determined by one of ordinary skill in the art. The determination of such values will depend at least in part on the manner in which the values are measured or determined, such as the limitations of the measurement system, i.e., the accuracy required for a particular purpose. For example, the term "about" may mean within 1 or a standard deviation of 1 or more, according to practice in the art. Alternatively, when referring to numerical values, the term "about" may mean ± 20%, typically ± 10%, typically ± 5%, more typically ± 1% of the numerical value. In general, however, where a particular value is described in the application and claims, unless otherwise specified, the term "about" means within an acceptable error range for the particular value, typically within one standard deviation.
In a particular aspect of the invention, the high affinity or selective CXCR4 binding peptide conjugate ("PC") has the formula:
Figure BDA0003765213480000091
or a pharmaceutically acceptable salt thereof,
wherein:
a is 0 or 1;
b is an integer between 1 and 4;
AA 1 and the sulfur atom to which it is attached is 3-mercaptopropionic acid, optionally substituted cysteine, or optionally substituted homocysteine;
AA 1 and the sulfur atom to which it is attached is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine;
AA 2 and the sulfur atom to which it is attached is cysteine or homocysteine;
Ar 1 is an optionally substituted aryl group;
X 1 is Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or Lys (iPr);
AA 3 each is independently Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), Lys (iPr);
and wherein AA 1 、X 1 、AA 2 Or AA 3 Comprises a moiety of formula (la):
-L 1 –Q,
wherein
L 1 Is a polymer linker having a functional group for linking Q, wherein L 1 Comprising from about 2 to about 20 monomers or copolymers;
q is absent, is AA 4 Or a structural formula: - [ AA 4 -Y 1 -L 2 ] c -Y 2 A part of the group-Z,
wherein
c is 0 or 1;
AA 4 is an amino acid or a derivative thereof;
Y 1 is amino acid AA 4 A side chain functional group of (1);
L 2 is a non-polymeric linker;
z is a compound of pharmaceutical value;
Y 2 is a functional group or L of the compound having a medicinal value 2 A functional group of (a); wherein, AA 1 、AA 2 、X 1 、AA 3 And AA 4 With a positron-emitting radioisotope, radiometal isotope, deuterium, tritium, or combinations thereof, 13 C、 14 C、 18 O or a combination thereof.
"pharmaceutically acceptable salts" of a compound refer to salts that are pharmaceutically acceptable and possess the desired pharmaceutical activity of the parent compound. Such salts include: (1) acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or from organic acids, such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo [2,2,2] -oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, dodecylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, hexadiene diacid, and the like; or (2) a salt formed when an acidic proton present in the parent compound is replaced with a metal ion (e.g., an alkali metal ion, an alkaline earth metal ion, or an aluminum ion); or a salt formed by complexing with an organic base (e.g., ethanolamine, diethanolamine, triethanolamine, aminobutanetriamine, N-methylglucamine, etc.).
It will be understood that, when referring to an amino acid, the term "optionally substituted" means that the side chain functionality of the amino acid or the-amino group of the amino acid may be substituted, for example, with an alkyl group (e.g. C) 1 -C 10 Alkyl) or functional group protecting groups, and the like. For example, for lysine or other amino acids having nitrogen-containing heteroatom side chains, optionally substituted or "derivatives thereof" include those amino acids substituted with alkyl groups, nitrogen protecting groups (e.g., acyl-containing moieties), and/or haloalkyl groups (e.g., trifluoromethyl, etc.). For cysteine and other sulfur heteroatom-containing amino acids (including homocysteine and penicillamine), the terms "optionally substituted" and "derivatives thereof" may include those optionally substituted on the sulfur atom or the alpha-amine functional group, such as those with alkyl, thiol, or amine protecting groups, and the like. In general, when referring to AA 1 And AA 4 The term optionally substituted refers to substitution on the alpha-amine group. Exemplary substitutions on the alpha-amine group include, but are not limited to, alkyl and amine protecting groups, as well as those other substituents known to those skilled in the art. "protecting group" refers to a moiety that, when attached to a reactive group in a molecule, reduces or prevents this reactivity. Examples of protecting Groups are given in T.W.Greene and P.G.M.Wuts, Protective Groups in Organic Synthesis,3 rd edition,JohnWiley&Sons, New York,1999, and Harrison et al, Compendium of synthetic Organic Methods, Vols.1-8(John Wiley and Sons,1971-1996), the entire contents of which are incorporated herein by reference.
AA 4 Is an amino acid or a derivative thereof, wherein AA 4 Having pendant functional groupsY 1 . The presence of a pendant functional group allows the linker L to be 2 To amino acid AA 4 . It is to be understood that when the functional group Y is 1 Is connected to L 1 In this case, the number of hydrogen atoms bonded thereto is reduced by one. For example, in lysine, the side chain functionality is-NH 2 Therefore when it is not connected to L 2 When, Y 1 is-NH 2 When it is connected to L 2 When, Y 1 is-NH-. Thus, it should be understood that the connection at Y is omitted for simplicity only 1 On or present in Y 1 The number of hydrogen atoms in (c). Furthermore, the side chain functionality of any amino acid can be readily altered by one skilled in the art. For example, one skilled in the art can use known reaction procedures to change the hydroxyl side chain functionality of serine to amine, carboxylic acid, or other functional groups. In addition, the amine side chain functionality of lysine (i.e., -NH) 2 ) Can be changed into azide, -NH-N 3 Hydroxyl, amide, phosphate, thiol, and the like.
In a particular embodiment, X 1 Is labeled with an isotope. In another embodiment, AA 3 Is labeled with an isotope.
In some embodiments, X 1 And/or AA 3 Is labeled with an isotope. Isotopically labeled compounds of structural formula I are useful for imaging and/or diagnostic purposes, among others. Exemplary isotopically labeled X 1 And/or AA 3 Including those containing positron-emitting radioisotopes, e.g. 34 Cl、 45 Ti、 51 Mn、 61 Cu、 63 Zn、 68 Ga、 11 C、 13 N、 15 O and 18 F. when radioisotopes are used, such radioisotopes are typically complexed to X by complexing or complexing agents 1 And/or AA 3 . Suitable complexing or coordinating moieties are well known to those skilled in the art. Other useful isotopic labels include, but are not limited to, deuterium (D), tritium (T), 13 C、 14 C、 18 O or a combination thereof.
Another example of a useful imaging agent includes a radioactive metal isotope that is coordinated (i.e., chelated) with a chelating group. Particularly useful radioactive metal isotopes include technetium, rhenium, gallium, gadolinium, indium, copper, and combinations thereof. Suitable chelating groups for particular radioactive metal isotopes are well known to those skilled in the art. For example, ferrocene and its derivatives, ethylenediaminetetraacetic acid ("EDTA"), derivatives thereof, peptidyl moiety Dap-Asp-Cys and derivatives thereof (see U.S. patent No.7128893), and others known in the art.
Yet another example of a useful imaging agent includes a contrast agent. Contrast agents are widely used, for example in Magnetic Resonance Imaging (MRI). A variety of contrast agents are known to those skilled in the art, including gadobenate, gadobutrol, gadodiamide, gadofosveset (gadofosveset), gadopentetate solution (gadopentetate), gadoterate (gadoterate), gadoteridol, gadofoscamine (gadoversetamide), gadoxetate (gadoxetate), and ferric oxide.
Another example of useful imaging agents include fluorescent dyes such as Fluorenylmethyloxycarbonyl (FMOC) and its derivatives, AlexaFluor dyes, Oregon Green dyes, fluorescein, BODIPY (borodipyrrolidine) dyes, cyanine dyes, rhodamine dyes, DyLight dyes, and texas red.
In other embodiments, X 1 And/or AA 3 Comprising (e.g., attached) or linked) a diagnostic agent, such as an imaging agent, an isotopic agent, or a radioactive agent. In a particular embodiment, X 1 And/or AA 3 Is labeled with an isotope. In some cases, X 1 And/or AA 3 Is lysine substituted with an isotopically labeled side chain. As used herein, the term "side-chain" or "side-chain" when referring to an amino acid is used to describe the "R" group in the following structural formula: h 2 N–CH(R)–CO 2 H. The side chains of a given amino acid are well known to those skilled in the art, e.g., the side chain of glycine or "R" is H, and for serine is of the formula-CH 2 A portion of OH, etc. In other cases, X 1 And/or AA 3 Is lysine, in which the side chain functional group amine is substituted with an isotopically labeled alkyl group. The term "isotopically-labeled alkyl" as used herein means that a carbon or hydrogen atom in the alkyl group is replaced byBy corresponding isotopes, e.g. carbon atoms by 13 C and/or 14 C, and hydrogen atoms are replaced by deuterium or tritium. In some cases, the amine nitrogens of the lysine side chain functional groups are used 13 And (4) labeling with N isotope. In other embodiments, with 18 The carbonyl oxygen of the amino acid is marked by an O isotope. In other embodiments, amino acid X 1 And/or AA 3 For one or more carbon atoms in 13 C or 14 And C isotope labeling. In other embodiments, with 14 N isotope labeled amino acid X 1 And/or AA 3 The alpha-amino functional group of (a). In other embodiments, amino acid X 1 And/or AA 3 The alpha-amino function of (a) may be substituted with an isotopically-labelled group, for example an isotopically-labelled carbonyl group (e.g. isotopically-labelled 18 O、 13 C、 14 C-labeled carbonyl) or isotopically labeled alkyl (e.g., D, T, 13 C or 14 C)。
In one embodiment, X 1 And/or AA 3 Is lys (iPr) or D-lys (iPr) wherein one or more, typically two or more, typically three or more, more often four or more, more often five or more and most often at least six hydrogen atoms on the iPr group are labelled (i.e. substituted) with deuterium or tritium isotopes.
As shown in structural formula I, in one embodiment, the peptidyl group is formed by using a functional group "Y" present on a compound of pharmaceutical value 2 "or in a linker L 2 Functional group "Y" of 2 "a compound of pharmaceutical value linked to structural formula Z. Such connections are well known and can be readily implemented by those skilled in the art. For example, when the functional group Y 2 Is a joint L 2 In the case of part (a), the compound Z of pharmaceutical interest is modified in such a way that a detachable group or other suitable bond-forming agent is present in Z, for example if Y 2 Is L 2 Can be used to substitute halides or other suitable detachable groups (e.g. methanesulfonic acid, toluenesulfonic acid, etc.), or to attach to the carbonyl group present in a compound Z of pharmaceutical value, in the form of the respectiveTo form amine or amide bond. Similarly, if Y is 2 Is a hydroxyl group, it may be used to replace a halide or other detachable group, or to attach to a carbonyl group present in a compound of pharmaceutical value, to form an ether or ester, respectively. In a similar manner, if the functional group Y 2 In the pharmaceutically valuable compound Z, Y 2 Can be used for connecting joints L 2 . For example, by linking a hydroxyl group or an amino group present on a compound of medical value to a linker L present on a non-polymer 2 The carbonyl group of (a). In this way, it is possible to use compounds of medical value and linkers L 2 Respectively forms ester bonds or amido bonds between the two. After reading this disclosure, one of ordinary skill in the art will readily envision a use for Y 2 Other suitable functional groups of (a).
In still other embodiments, L 2 Is selected from-CH 2 –C(=O)–、–NH–CH 2 –、–(CH 2 ) n –C(=O)–NH–CH 2 –)、–(CH 2 ) n –NH–C(=O)–CH 2 -, or- (CH) 2 ) n -a non-polymeric crosslinker of formula (la) wherein n is an integer between 1 and 6. In a particular embodiment, L 2 is-CH 2 -C (═ O) -and Y 2 Is O.
In formula I, amino acid AA 4 Is a compound having a functional group-Y 1 Amino acids of the side chain of (1). This allows the joint L to be connected 2 To the hetero atom Y 1 . Thus, the amino acid AA is suitable 4 Including but not limited to serine, cysteine, lysine, arginine, aspartic acid, glutamine, glutamic acid, histidine, proline, threonine, tryptophan, tyrosine, and the like, as well as homologs or derivatives thereof, e.g., homocysteine, homoserine. In addition, AA 4 It may also be a non-proteinogenic, synthetic or "unnatural amino acid", i.e. an amino acid which does not occur naturally but is known to the person skilled in the art, such as ornithine, 1, 4-diaminobutyric acid (Dab), 1, 3-diaminopropionic acid (Dap), penicillamine and the like. In a particular embodiment, AA 4 Is cysteine,Homocysteine or penicillamine. In this case, Y 1 Is sulfur (e.g., a mercaptan). In another embodiment, AA 4 Is serine, in this case Y 1 Is oxygen (e.g., hydroxyl).
Unless otherwise indicated or the context requires otherwise, the amino acids disclosed herein may be in the (D) -or (L) -stereo configuration. Indeed, in some embodiments, one or more amino acids are in the (D) -configuration. In this way, the in vivo stability of the compounds of structural formula I may be greatly enhanced or increased. Typically, the amino acids of the invention are proteinogenic amino acids, unless otherwise specified.
In other embodiments, L 1 Is a polymer linker comprising from about 2 to about 20, typically from about 2 to about 15, typically from about 2 to about 10, more often from about 2 to 8, and most often from about 5 to about 8 monomers or copolymers. Copolymer refers to a linker having two or more different monomer units. For example, the copolymer may be between ethylene glycol-propylene glycol units; ethylene glycol and vinyl alcohol units; a polymer formed between ethylene glycol and vinyl acetate units; and other copolymers well known to those skilled in the art. It is understood that L 1 Two terminal units (e.g., attached to AA) 3 And is attached to AA 4 Another end unit of) is functionalized so that it can be easily attached to AA 3 And AA 4 . For example, when L is 1 Is polyethylene glycol ("PEG"), attached to AA 3 One end of PEG of (A) may be functionalized as an amine so that it reacts with AA 3 Forming an amide bond. On the other hand, if attached to AA 3 The PEG of (2) is kept as a hydroxyl group at the end, then it is reacted with AA 3 An ester bond is formed. Similarly, if attached to AA 4 The hydroxyl group of PEG of (A) is oxidized to a carboxylic acid moiety, then it is reacted with AA 4 The alpha-amino function of (a) forms an amide group. Or, when attached to AA 4 When the hydroxyl group of PEG of (A) is substituted with a detachable group (e.g., toluenesulfonic acid, methanesulfonic acid, halide, etc.), it may be reacted with AA by, for example, a displacement reaction 4 The alpha-amino function of (a) forms an amino bond. In this way, L can be formed using a variety of methods 1 Attached to AA 3 And AA 4 . In a particular embodiment, L 1 Is a polyethylene glycol in which AA is attached 3 Is amino at the end of (A), thereby reacting with AA 3 Forming an amide bond. In another embodiment, L 1 Is a polyethylene glycol in which AA is attached 4 Is oxidized to a carboxylic acid or similar functional group, thereby reacting with the amino acid AA 4 The alpha-amino function of (a) forms an amide bond.
In some embodiments, a is 1. In a particular embodiment, AA 3 Is lysine or a derivative thereof (e.g., the amino group of the side chain is alkylated, acylated, or substituted with an amine protecting group), optionally isotopically labeled. In some embodiments, the pendant amino functional group is alkylated with an alkyl group optionally having one or more deuterium or tritium atoms on the alkyl group in place of hydrogen. In some embodiments, the side chain amino functional group is acylated, which is optionally isotopically labeled, e.g., isotopically labeled, carbonyl oxygen (e.g., isotopically labeled) 18 O) and/or isotopically-labeled carbonyl carbons (e.g. 13 C or 14 C) And the like. In a particular embodiment, AA 3 Is lysine with an isopropyl group attached to a side chain amino group. In a particular embodiment, the isopropyl group is optionally isotopically labeled, for example, with multiple deuterium and/or tritium atoms.
In some embodiments, a is 0. In this embodiment, the compound has a disulfide bond to form a ring structure.
In other embodiments, AA 3 Is lysine or a derivative thereof (e.g., the amino group of the side chain is alkylated, acylated, or substituted with an amine protecting group), optionally isotopically labeled. In some embodiments, the pendant amino functional group is alkylated with an alkyl group optionally having one or more deuterium or tritium atoms on the alkyl group in place of hydrogen. In some embodiments, the side chain amino functional group is acylated, which is optionally isotopically labeled, such as isotopically labeled carbonyl oxygen (e.g., isotopically labeled carbonyl oxygen) 18 O) and/or isotopically-labeled carbonyl carbons (e.g. 13 C or 14 C) And the like. In a particular embodiment, AA 3 Is lysine having an isopropyl group attached to a side chain amino groupAnd (4) acid. In a particular embodiment, the isopropyl group is optionally isotopically labeled, for example, with multiple deuterium and/or tritium atoms.
In other embodiments, AA 2 And the sulfur atom to which it is attached is cysteine, homocysteine or penicillamine. Generally, AA 2 And the sulfur atom to which it is attached is cysteine or homocysteine. In a specific embodiment, AA 2 And the sulfur atom to which it is attached is cysteine.
In yet other embodiments, AA 1 And the sulfur atom to which it is attached is 3-mercaptopropionic acid (3-MPA), cysteine, homocysteine, or penicillamine. Generally, AA 1 And the sulfur atom to which it is attached is 3-MPA, cysteine or homocysteine. In a specific embodiment, AA 1 And the sulfur atom to which it is attached is cysteine.
In other embodiments, Ar 1 Is phenyl. -S-CH 2 The group can be positioned 1, 2-of the phenyl group; 1, 3-; or 1, 4-bit. In a particular embodiment, -S-CH 2 The group is positioned in the 1, 2-position, i.e. the phenyl group is 1, 2-disubstituted. In other embodiments, Ar 1 Is a phenyl group having one, two, three or four, typically one, two or three, typically one or two, and most typically one substituent. Is suitable for Ar 1 Exemplary substituents of (A) include, but are not limited to, halides (e.g., F, Cl, I, or Br), C 1 -C 10 Alkyl (e.g., methyl, ethyl, t-butyl, isopropyl, etc.), C 1 -C 10 Haloalkyl (e.g. -CF) 3 Etc.), nitro, nitroso, -CO n R (where n is 1 OR 2 and R is hydrogen OR alkyl), cyano, -OR (where R is H, alkyl, carbonyl, etc.), and other electron donating OR electron withdrawing groups known to those skilled in the art.
In yet other embodiments, linker L 2 Comprising the ability to release Y in vivo 2 -a functional group of Z. In this way, Y 2 the-Z moiety is released in vivo, thereby exerting its therapeutic activity. Can release Y 2 Suitable functions of-ZThe groups depend on the attachment to the linker L 2 Part Y of 2 The nature of the functional group(s) above. For example, when the functional group on Z is hydroxy (i.e., Y) 2 is-OH) or amino (Y) 2 is-NH 2 ) When L is 2 The functional group in (A) may be a carboxylate, so that the compounds and linkers L having medical value 2 Respectively form ester bonds or amido bonds between the two. If the functional group on the compound has medical value (i.e. "Y 2 ") is a carboxylic acid, then L 2 The corresponding functional group on (a) may be a hydroxyl group or an amino group, forming an ester bond or an amide bond, respectively. L is well known to those skilled in the art 2 Other suitable functional groups capable of releasing compounds of medical value in vivo include disulfide bonds, ester bonds, mercaptomaleimide bonds, and the like.
In other embodiments, the compound of pharmaceutical value is a therapeutic agent. Suitable therapeutic agents include those known to those skilled in the art for the treatment of cancer, autoimmune diseases (e.g., rheumatoid arthritis), viral infections (e.g., HIV infection), and the like. Exemplary therapeutic agents useful in the compounds of the present invention include, but are not limited to, altretamine; an asparaginase enzyme; bleomycin; busulfan; carboplatin; carmustine; chlorambucil; cisplatin; cladribine; cyclophosphamide; cytarabine; (ii) an azelamide; diethylstilbestrol; eribulin, ethinylestradiol; etoposide; mitomycin; o-chlorobenzene-p-chlorobenzene dichloroethane; mitoxantrone; paclitaxel; pentostatin; bromopropylpiperazine; mithramycin; prednisone; procarbazine; streptozotocin; tamoxifen; (ii) teniposide; vinblastine; vincristine, daunorubicin, doxorubicin, docetaxel, irinotecan, methyl-auristatin E, mertansine, SN-38, tesirine, telithsin, vinblastine and analogs or derivatives thereof, HIV protease inhibitors, HIV fusion inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV entry inhibitors, and therapeutic agents for autoimmune diseases.
L 2 Can be any biocompatible bifunctional linker, such as polyethylene glycol (PEG), e.g., H 2 N–CH 2 CH 2 –(PEG)m–CH 2 CH 2 –COOH、HOOC–CH 2 CH 2 –(PEG)m–CH 2 CH 2 -COOH or H 2 N–CH 2 CH 2 –(PEG)m–CH 2 CH 2 –NH 2 Natural and non-natural amino acids or in the form of polyamic acids (PAA), where m is an integer from 0 to 100, typically from 1 to 50, typically from 1 to 25, more typically from 1 to 10. Generally, when L is 2 In the case of polymers (e.g., PEG, PAA), the total number of monomers in the chain is from about 2 (i.e., monomers) to about 20, typically from about 2 to about 15, typically from about 3 to about 10, and most typically from about 4 to about 6.
In other embodiments, the compound of medical value is a diagnostic or imaging agent, such as a radioactive agent, a fluorescent agent, and the like. Such imaging agents are well known to those skilled in the art. For example, contrast agents for magnetic resonance imaging agents, ultrasound contrast agents and radiological contrast agents. See, for example, en. wikipedia. org/wiki/Contrast agent.
In addition, combinations of the various groups described herein may form other embodiments. In this way, a variety of compounds are presented in the present invention. By combining the various groups described herein in different ways, for example, compounds of structural formulae IA and IB are included within the scope of the present invention:
Figure BDA0003765213480000161
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、L 1 Q and Ar 1 As defined herein, and
Figure BDA0003765213480000162
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、AA 4 、Y 1 、Y 2 、L 1 、L 2 Z and Ar 1 As defined herein.
Some specific examples of compounds of the present invention include, but are not limited to, compounds of formula A, compounds of formula B, compounds of formula C, compounds of formula D, compounds of formula E, compounds of formula F, compounds of formula G, compounds of formula H, compounds of formula I, compounds of formula J, compounds of formula K, compounds of formula L, compounds of formula M, compounds of formula N, compounds of formula O, and compounds of formula P, each of which may also be labeled with deuterium (e.g., compounds of formulae A-D12, B-D12, C-D12, etc.) or tritium (e.g., compounds of formulae A-T12, B-T12, C-T12, …, P-T12). The examples section provides the synthesis of these compounds.
It is to be understood that in the present invention, combinations of the different embodiments described herein form further preferred embodiments. For example, one particular embodiment is described herein as "a" being 0, and another embodiment is described herein, wherein AA is 1 Is homocysteine. Thus, the combination of these two embodiments provides compounds of structural formulae I, IA and IB wherein a is 0, AA 1 Is homocysteine.
Another aspect of the invention provides a diagnostic kit comprising a high affinity CXCR4 selective binding peptide conjugate described herein, wherein the compound of medical value is a diagnostic agent.
In another aspect of the present invention, there is provided a composition comprising a compound of structural formula I and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier may include diluents, excipients, flavoring agents, adjuvants, binders, stabilizers, coloring agents, or combinations thereof. In general, a "pharmaceutically acceptable carrier" refers to any excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and not biologically or otherwise undesirable, and includes excipients that are acceptable for veterinary use as well as human medicine that is of medical value.
The present invention includes pharmaceutical compositions comprising at least one compound of the present invention, or individual isomers, racemates or non-racemic mixtures of isomers or pharmaceutically acceptable salts or solvates thereof, together with at least one pharmaceutically acceptable carrier, and optionally other therapeutic and/or prophylactic ingredients.
In general, the compounds of the invention are administered in therapeutically effective amounts by any acceptable mode of administration that provides for agents of similar use. Suitable dosage ranges are typically from 1 to 500mg per day, typically from 1 to 100mg per day, typically from 1 to 30mg per day, depending on a number of factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound used, the route and form of administration, the indication for which administration is being made, and the preference and experience of the relevant physician. One of ordinary skill in the art of treating such diseases will generally be able to determine, without undue experimentation and relying on personal knowledge and the disclosure of this application, therapeutically effective amounts of the compounds of the present invention.
In general, the compounds of the invention are administered as pharmaceutical formulations, including those suitable for oral (including buccal and sublingual), rectal, nasal, topical, pulmonary, vaginal or parenteral (including intramuscular, intraarterial, intrathecal, subcutaneous and intravenous) administration, or in a form suitable for administration by inhalation or insufflation. Typical modes of administration are generally oral, and convenient daily dosage regimens may be adjusted to acceptable levels.
One or more compounds of the invention, together with one or more conventional adjuvants, carriers or diluents, may be used in the form of pharmaceutical compositions and unit dosages. The pharmaceutical compositions and unit dosage forms may be comprised of conventional ingredients in conventional proportions, with or without additional active compounds or ingredients (principle), and the unit dosage forms may contain any suitable effective amount of the active ingredient corresponding to the intended daily dosage range to be employed. The pharmaceutical compositions may be employed as solids, such as tablets or filled capsules, semisolids, powders, sustained release formulations, or liquids, such as solutions, suspensions, emulsions, elixirs, or orally filled capsules; or in the form of suppositories for rectal or vaginal administration; or in the form of a sterile injectable solution for parenteral use. Thus, formulations containing about one (1) milligram of active ingredient per tablet or, more broadly, about 0.01 to about one hundred (100) milligrams of active ingredient are suitable representative unit dosage forms.
The compounds of the present invention may be formulated into a variety of oral dosage forms. Pharmaceutical compositions and dosage forms may contain one or more compounds of the invention or pharmaceutically acceptable salts thereof as the active ingredient. The pharmaceutically acceptable carrier may be a solid or a liquid. Solid formulations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances and may also act as a diluent, flavoring agent, solubilizer, lubricant, suspending agent, binder, preservative, tablet disintegrant, or encapsulating material. In powders, the carrier is typically a finely divided solid which is in admixture with the finely divided active ingredient. In tablets, the active ingredient is usually mixed with the carrier having the necessary binding capacity in suitable proportions and compacted in the shape and size desired. Powders and tablets preferably contain from about one (1)% to about seventy (70)% of the active compound. Suitable carriers include, but are not limited to, magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, gum tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low melting wax, cocoa butter, and the like. The term "preparation" is intended to include the formulation of the active compound with encapsulating material as a carrier, providing a capsule in which the active ingredient, with or without a carrier, is surrounded by a carrier which is in association therewith. Similarly, cachets and lozenges are also included. Tablets, powders, capsules, pills, cachets, and lozenges can be in solid form suitable for oral administration.
Other forms suitable for oral administration include liquid form preparations including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions or solid form preparations which are converted to liquid form prior to use. Emulsions may be prepared in solution, for example in aqueous propylene glycol, or may contain emulsifying agents, for example lecithin, sorbitan monooleate, or acacia. Aqueous solutions may be prepared by dissolving the active ingredient in water and adding suitable colorants, flavors, stabilizers, and thickening agents. Aqueous suspensions may be prepared by dispersing the comminuted active ingredient in water using viscous materials, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents. Solid preparations which are intended to be converted into liquids immediately before use include solutions, suspensions and emulsions, which may contain, in addition to the active ingredient, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
The compounds of the invention may also be formulated for parenteral administration (e.g., by injection, e.g., bolus injection or continuous infusion) and may be presented in unit dosage form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, for example, as solutions in aqueous polyethylene glycol. Examples of oily or nonaqueous carriers, diluents, solvents or vehicles include propylene glycol, polyethylene glycol, vegetable oils (e.g., olive oil) and injectable organic esters (e.g., ethyl oleate), and may contain formulatory agents such as preservatives, wetting, emulsifying or suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of a sterile solid or by lyophilization from solution, before use in association with a suitable carrier, e.g., sterile pyrogen-free water.
The compounds of the invention may be formulated as ointments, creams or lotions or as a transdermal patch for topical application to the epidermis. For example, ointments and creams may be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Emulsions may be formulated with an aqueous or oil base and typically also contain one or more emulsifying, stabilizing, dispersing, suspending, thickening or coloring agents. Formulations suitable for topical administration in the oral cavity include lozenges comprising the active agent, usually sucrose and acacia or tragacanth, in a flavored base; lozenges comprise the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
The compounds of the invention may be formulated for administration as suppositories. A low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, is first melted and the active ingredient is dispersed homogeneously, for example by stirring. The molten homogeneous mixture is then poured into suitably sized moulds, cooled and solidified.
The compounds of the invention may also be formulated for vaginal administration. In addition to the active ingredient, an embolus, tampon, cream, gel, paste, foam or spray containing a suitable carrier as known in the art.
The compounds of the invention may be formulated for nasal administration. The solution or suspension is applied directly to the nasal cavity by conventional means, for example, using a dropper, pipette or nebulizer. These formulations may be provided in single or multiple dose forms. In the case of the latter dropper or pipette, this may be achieved by the patient administering an appropriate predetermined volume of solution or suspension. In the case of spraying, this can be achieved, for example, by means of a metered atomizing spray pump.
The compounds of the invention may be formulated for aerosol administration, particularly to the respiratory tract, including intranasal administration. The particle size of the compound is typically small, for example five (5) microns or less. Such particle sizes may be obtained by methods known in the art, for example by micronisation. The active ingredient is provided in a pressurized pack containing a suitable propellant, for example chlorofluorocarbon (CFC), such as dichlorodifluoromethane, trichlorofluoromethane or dichlorotetrafluoroethane, or carbon dioxide or other suitable gas. The aerosol may also conveniently comprise a surfactant, for example lecithin. The dose of medicament may be controlled by a metering valve. Alternatively, the active ingredient may be provided in the form of a dry powder, for example, a powder mixture of the compound in a suitable powder base, such as lactose, starch derivatives (e.g., hydroxypropylmethyl cellulose and polyvinyl pyrrolidone (PVP)). Powder carriers typically form gels in the nasal cavity. The powder compositions may be presented in unit dosage form, for example, in capsules or cartridges, such as gelatin or blister packs, from which the powder may be administered by means of an inhaler.
When desired, the formulations may be prepared using enteric coatings suitable for sustained or controlled release administration of the active ingredient. For example, the compounds of the present invention may be formulated in a transdermal or subcutaneous drug delivery device. These delivery systems are advantageous when sustained release of the compound is required or desired, and when patient compliance with a treatment regimen is critical. The compounds in transdermal delivery systems are often attached to a skin adhesive solid carrier. The target compound may also be combined with a penetration enhancer such as azone (1-dodecylazacycloheptan-2-one). Sustained release drug delivery systems can be implanted subcutaneously by surgery or injection. Subcutaneous implants encapsulate the compound in a lipid-soluble membrane (e.g., silicone rubber) or a biodegradable polymer (e.g., polylactic acid).
Pharmaceutical formulations are generally in unit dosage form. In this form, the preparation is generally subdivided into unit doses containing appropriate quantities of the active ingredient. The unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparation, such as packeted tablets, capsules, and powders in vials or ampoules. In addition, the unit dosage form can be a capsule, tablet, caplet, or lozenge itself, or any suitable number of such packaged forms.
Other suitable pharmaceutical carriers and formulations thereof are described in Remington: the Science and practice of pharmacy 1995, E.W. Martin, Mack Publishing Company, 19 th edition, Easton, Pa.
The active ingredients may be presented as pharmaceutical compositions when it is possible to use therapeutically effective amounts of the compounds of structural formula I and pharmaceutically acceptable salts thereof as starting chemicals in therapy. Accordingly, the present invention further provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of structural formula I, or a pharmaceutically acceptable salt or prodrug thereof, in combination with one or more pharmaceutically acceptable carriers, diluents or excipients. When applied to a combination, the term refers to the combined amounts of the active ingredients that produce a therapeutic effect, whether administered in combination, sequentially or simultaneously. Compounds of structural formula I and pharmaceutically acceptable salts thereof are described above. The carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. According to another aspect of the present invention there is also provided a process for the preparation of a pharmaceutical formulation comprising mixing a compound of structural formula I or a pharmaceutically acceptable salt or prodrug thereof with one or more pharmaceutically acceptable carriers, diluents or excipients.
When the compositions of the present invention comprise a combination of a compound of the present invention and one or more additional therapeutic or prophylactic agents, the compound and additional agent are typically present at dosage levels of about 10% to 150%, more typically about 10% to 80% of the dosage normally administered in monotherapy.
Another aspect of the invention provides a method of imaging cancer cells in a patient comprising administering to the patient an effective imaging amount of a high affinity CXCR4 selective binding peptide conjugate of structural formula I, a compound of pharmaceutical value as an imaging agent, and imaging cancer cells in the patient using an imaging device. The imaging device used depends on the nature of the imaging agent of the compound of formula I. For example, if the imaging agent is a positron-emitting radioisotope, the imaging device used is a PET scan, and when the compound of medical value is a contrast, the imaging device may be a computed tomography imaging device or an MRI device. When the pharmaceutically valuable compound is a radioisotope, the imaging device may be an x-ray machine or other similar device.
In one particular aspect of the invention, a method of treating cancer in a patient is provided. The method comprises administering to a cancer patient a therapeutically effective amount of a compound of structural formula I wherein the compound of pharmaceutical value is a cancer drug or a pharmaceutical composition comprising a compound of structural formula I wherein the compound of pharmaceutical value is a cancer drug.
Another particular aspect of the invention provides a diagnostic or imaging kit comprising a high affinity CXCR4 selective binding Peptide Conjugate (PC) of structural formula I, wherein the compound of medical value is a diagnostic agent or an imaging agent, respectively.
Another particular aspect of the invention provides a method of treating a patient suffering from rheumatoid arthritis, pulmonary fibrosis, HIV infection or cancer. The method comprises administering to a patient in need of treatment a therapeutically effective amount of a compound of structural formula I. In this method, compounds of formula I having medical value are therapeutic agents useful in treating the particular clinical condition being treated. Some cancers that may be treated using the compounds of the present invention include, but are not limited to, breast cancer, pancreatic cancer, melanoma, prostate cancer, renal cancer, neuroblastoma, non-hodgkin's lymphoma, lung cancer, ovarian cancer, colorectal cancer, multiple myeloma, glioblastoma multiforme, and chronic lymphocytic leukemia.
Other objects, advantages and novel features of the present invention will become apparent to those skilled in the art upon examination of the following examples, which are not intended to limit the invention. In these examples, procedures that are constructively simplified to practice are described in the present time and procedures performed in the laboratory are described in the past time.
Examples
The following abbreviations are used: ac: acetyl; boc: a tert-butoxycarbonyl group; BOP: (benzotriazole-1-acyloxy) -tris (dimethylamino) phosphonium hexafluorophosphate; bz: a benzoyl group; bzl: a benzyl group; dab 1, 4-diaminobutyric acid; dap: 1, 3-diaminopropionic acid; DCC: dicyclohexylcarbodiimide; DCM: dichloromethane; DIC: diisopropylcarbodiimide; DIEA: diisopropylethylamine; DMAP: 4- (N, N-dimethylamino) pyridine; DMF: n, N-dimethylformamide; DMSO (dimethylsulfoxide): dimethyl sulfoxide; EDT 1, 2-ethane-dithiol; et: an ethyl group; fmoc: 9-fluorenylmethoxycarbonyl; HATU: n- [ (dimethylamino) -1H-1, 2, 3-triazolyl [4, 5-b ] pyridin-1-ylmethylene ] -N-methylformamide hexafluorophosphate nitroxide; HBTU: O-benzotriazolyl-N, N' -tetramethylurea hexafluorophosphate; HCTU: 1H-benzotriazole 1- [ bis (dimethylamino) methylene ] -5-chloro-3-oxide hexafluorophosphate; HOBt: hydroxybenzotriazole; hCys: homocysteine; iPr: an isopropyl group; IPA: isopropyl alcohol; me: a methyl group; mmt: 4-methoxytriphenyl; mpa: 3-mercaptopropionic acid; 2 Nal: 2-naphthylalanine; 1 Nal: 1-naphthylalanine; NMM: n-methylmorpholine; NMP: n-methyl pyrrolidone; orn: ornithine; pbf: 2,2,4,6, 7-pentamethyldihydrobenzofuran-5-sulfonyl; PBS: phosphate buffer solution; PyBOP: (benzotriazol-1-yloxy) -tris (pyrrolidinyl) -phosphonium hexafluorophosphate; PyBrOP: trispyrrolidinylphosphonium bromide hexafluorophosphate; tBu: a tertiary butyl group; TFA: trifluoroacetic acid; TFE: trifluoroethanol; THF: tetrahydrofuran; and (3) TIS: triisopropylsilane; trt: triphenyl radicals; mini-PEG 6: 6-mers of ethylene glycol; all common amino acids are represented by three letter symbols or other designated symbols.
Mass Spectrometry (MS) analysis: the preparation of the compounds of the present invention described in the following examples is intended to be illustrative and not limiting. In these examples, the observed molecular weights are all reported as deconvoluted (de-convolved) values. The deconvolution value is obtained from the formula MW (observed value) ═ n (m/z) -n, where m/z represents a charged ion (positive ion mode), and n is the charge number of the specific substance. When multiple charged species are present in the mass spectrum, the observed molecular weights are reported as averages.
General methods for peptide synthesis, ring structure formation and salt exchange: peptides were chemically synthesized using solid phase peptide synthesis known in the art. The cyclic structure of these peptides is established by nucleophilic substitution using bis (halomethyl) aryl compounds in the presence of bases, typically 1.3 equivalents of bis (bromomethyl) aryl compounds, such as 15mM ammonium bicarbonate solution, for disulfides, by oxidation with air or iodine in the presence of acids, or for bis-thioether rings.
While the present invention illustrates the preparation of one particular peptide chain, other peptide chains within the scope of the present invention can be readily prepared using the disclosed methods, for example, commonly assigned U.S. patent application No.15/898434 filed on 2.17.2018 and 15/695862 filed on 9.5.2017, which are incorporated herein by reference in their entirety. In addition, other peptide chains within the scope of the invention may be readily prepared by those skilled in the art after reading this disclosure and the commonly assigned U.S. patent applications incorporated herein by reference.
Activation of paclitaxel-preparation of 2' -maleimidopaclitaxel:
Figure BDA0003765213480000231
to a 0 ℃ solution of 80mL DCM containing 500mg paclitaxel (0.6mmol) and 0.06mmol DMAP (9.2mg) was added a solution of 1.1mmol 2-bromoacetic acid (100.3mg) in 50mL DCM, followed by 3 equivalents DIC (0.3mL) with stirring. The reaction mixture was then slowly warmed to room temperature and stirred continuously at 30 ℃ for 1h to continue the coupling reaction. The crude 2' -bromoacetyl paclitaxel was purified and lyophilized, and then bound to the cyclized CXCR4 antagonist peptide (molecular weight-mass spectrum: observed 975.10; calculated 974.85).
Purification, salt morphology conversion and final product characterization: the final product was purified by reverse phase HPLC and further characterized by analytical HPLC and mass spectrometry. The peptide purified from reverse phase high performance liquid chromatography is typically in the form of trifluoroacetic acid (TFA). The salts are typically converted to more pharmaceutically valuable salt forms, such as acetic acid or hydrochloride salt forms. Conversion of the peptide in the TFA salt to the hydrochloride salt can be achieved by repeated lyophilization of the peptide in the TFA salt in dilute hydrochloric acid solution. To convert the peptide in the TFA salt to the acetate salt, the following method is generally used. The strong anion exchange resin (chloride form, degree of substitution 3mmol/g, water content 50%, 2g resin per g peptide) was first washed three times with ultrapure (milli Q) water, then three times with 1N NaOH solution for 5min each, then five times with ultrapure water for 5min each. The resin was further washed with 75% ethanol-water until a pH of around 7.4 was reached. The resin was treated three times with 10% acetic acid solution for 5min each time. The resin was then washed three times with 1% acetic acid solution for 5min each. The resin was prepared for salt conversion of the purified peptide.
The purified lyophilized peptide was dissolved in a 1% acetic acid solution and added to the resin prepared above. The mixture was stirred at room temperature or magnetically for 1 h. The supernatant was separated. The resin was washed three times with 1% acetic acid solution. The supernatant and the washing were mixed, filtered through a 0.22 μm membrane and lyophilized to give the peptide in acetate.
Example 1:synthesis of Compound A
Figure BDA0003765213480000241
Peptide chain assembly:Fmoc-Cys(Mmt)-Tyr(tBu)-Lys(iPr,Boc)-(D-Arg(Pbf))-2Nal-Gly-Cys(Mmt)-Lys(iPr,Boc)-(mini-PEG6)-Cys (Trt) -Sieber amide AM resin (SEQ ID NO:5)
Figure BDA0003765213480000242
Peptide chains were assembled by standard Fmoc chemistry using Sieber amide AM resin (Xi' an lanjiao Chemical Limited, west ann, china). Briefly, 40g of Sieber amide AM resin was swollen in 300mL DCM for 2h, then washed four times with DMF. Fmoc was removed at room temperature in 250mL of 20% piperidine in DMF for 20min and washed several times with DMF. Ninhydrin test was negative. The chain was assembled stepwise starting from Fmoc-Cys (Trt) -OH at the C-terminus of the linear peptide. Three equivalent protected amino acids Fmoc-Cys (Trt) -OH were activated with DIC/HOBt in 80mL DMF and coupled to Fmoc-depleted Sieber amide AM resin prepared above for 2h at room temperature. Ninhydrin test was negative. A mixture of 160mL acetic anhydride/DIEA/DCM was used to cap the unreacted amino groups at a volume ratio of 1:1:4 for 30 min. Subsequently, Fmoc was removed with 20% piperidine in 250ml dmf for 20 min. The following residues were coupled sequentially without capping: fmoc- (mini-PEG6) -OH, Fmoc-Lys (iPr, Boc) -OH, Fmoc-Cys (Mmt) -OH, Fmoc-Gly-OH, Fmoc-2Nal-OH, Fmoc-D-Arg (Pbf) -OH, Fmoc-Lys (iPr, Boc) -OH, Fmoc-Tyr (tBu) -OH and Fmoc-Cys (Mmt) -OH. After coupling the final residue Fmoc-Cys (Mmt), Fmoc protection was removed again for 20min using 250mL of 20% piperidine in DMF. N-terminal acetylation was performed at room temperature for 30min using 160mL of an acetic anhydride/DIEA/DMF mixture (1:1:4, v/v/v). The resin was then washed three times with 250mL DMF followed by three times with 250mL DCM and then dried under vacuum to give 81 g peptide resin containing the assembled linear peptide of interest. The dried resin was divided into several portions, and one portion (20 g) was used in the following operation.
Removal of Cys residue protection by Mmt and cleavage of partially protected linear peptide from peptide resin: 20 g of the above peptide resin was swollen in 300ml of LPCM for 30 min. To the swollen resin above was added 1000mL of cleavage/deprotection mixture (TFA/H) 2 O/EDT/TIS/DCM, 6:3:1.5:3:86.5, v/v) to remove Mmt protection of the Cys side chain. The mixture was stirred at room temperature for 20 min. Using at room temperatureThe deprotection and cleavage procedure was repeated for 20min with 1000mL of cleavage/deprotection mixture. After removing the solid resin by filtration, the cleavage solutions containing the partially deprotected linear peptide were combined and concentrated under vacuum using a rotary evaporator. The residue (crude peptide) was then lyophilized to give 9.5 g of partially deprotected crude peptide.
Cyclization of: to 2.2g of the above crude peptide solution in 110mL of DMF was added 20mL of MeCN containing 368mg of 1, 2-bis (bromomethyl) benzene. The solution was mixed well and then 1630mL MeCN and 440mL water were added. The pH of the reaction mixture was adjusted to pH 8-9 using 1M ammonium carbonate solution. The cyclization reaction was allowed to proceed for 1h at room temperature. The reaction was monitored by MS. The reaction mixture was then divided into six equal portions and freeze-dried.
Deprotection of the side chain of cyclized peptide: cleavage cocktail (TFA/EDT/TIS/H) was used 2 O/Thioanisole/phenol, 81.5mL TFA, 2.5mL EDT, 1.5mL TIS, 5.0mLH per 100mL solution 2 O, 5.0mL thioanisole and 5.0 grams phenol) in 10mL containers at 30 ℃ for 60 min. To the cleavage mixture was added 4 volumes of cold diethyl ether. The crude peptide precipitate was separated by centrifugation at 3000rpm for 2 min. The crude peptide precipitate was washed three times with diethyl ether. The crude peptide was purified on preparative HPLC to purity>90% and freeze-dried (MW-MS: observed 1719.55; calculated 1720.18).
Preparation of PDC-CXCR4 peptide-paclitaxel drug conjugate: 452mg of 2' -bromoacetyl paclitaxel prepared above and 905mg of cyclized CXCR4 peptide were dissolved in 100mL of MeCN/water (1:1, v/v). The solution is adjusted to pH 7-7.5 using 0.5M ammonium bicarbonate. At room temperature, MS confirmed that the coupling reaction was complete within half an hour. The final product was purified using a reverse phase preparative column Daisogel (50X 250mm, 8 μm); mobile phase-solvent a: 0.1% TFA water; solvent B: 0.1% TFA acetonitrile. Fractions containing the desired product MB1707 were combined and lyophilized (TFA salt). Salt exchange as described above gives the peptide in the form of acetate. The final peptide product was 99.61% pure by HPLC; MW calibration: 2614.12 Da; observed MW value is 2613.30 Da.
Example 2:isotopic labelling compoundsSynthesis of substance A-D12
Peptide chain assembly: Fmoc-Cys (Mmt) -Tyr (tBu) -Lys (Boc) - (D-Arg (Pbf) -2Nal-Gly-Cys (Mmt) -Lys (Boc) - (mini-PEG6) -Cys (Trt) -Sieber amide AM resin (SEQ ID NO: 7).
Figure BDA0003765213480000261
Peptide chains were assembled by standard Fmoc chemistry using Sieber amide AM resin (Xi' an lanjiao Chemical Limited, west ann, china). Briefly, 1.5 g of Sieber amide AM resin was swollen in 30mL DCM for 2h, then washed four times with 25mL DMF. Fmoc was removed at room temperature in 25mL of DMF containing 20% piperidine for 20min and washed several times with DMF. Ninhydrin test was negative. The chain was assembled stepwise starting from Fmoc-Cys (Trt) -OH at the C-terminus of the linear peptide. Three equivalents of the protected amino acid Fmoc-Cys (Trt) -OH (702mg) were activated by DIC (1mL)/HOBt (243mg) in 5mL of DMF and coupled overnight in a temperature controlled oven at 30 ℃ to Fmoc-removed Sieber amide AM resin prepared as described above. The reaction mixture was drained and the resin was washed several times with 25mL of DMF. Capping with 16mL of a mixture of acetic anhydride/DIEA/DCM at a volume ratio of 1:1:4 for 30min of unreacted amino groups. Ninhydrin test was negative. Fmoc was then removed for 20min using 20% piperidine in 25mL DMF.
The following residues were coupled sequentially without an end-capping step: fmoc- (mini-PEG6) -OH, Fmoc-Lys (Boc) -OH, Fmoc-Cys (Mmt) -OH, Fmoc-Gly-OH, Fmoc-2Nal-OH, Fmoc-D-Arg (Pbf) -OH, Fmoc-Lys (Boc) -OH, Fmoc-Tyr (tBu) -OH and Fmoc-Cys (Mmt) -OH. After coupling the final residue Fmoc-Cys (Mmt), Fmoc protection was removed again for 20min using 25mL of 20% piperidine in DMF. N-terminal acetylation was performed at room temperature for 30min using 16mL of an acetic anhydride/DIEA/DMF mixture (1:1:4, v/v/v). The peptide resin was then washed three times with 25mL DMF followed by three times with 25mL DCM, after which the peptide resin was dried under vacuum to give 2.8 g peptide resin containing the assembled target linear peptide.
Removal of Cys residue protection by Mmt and cleavage of partially protected linear peptide from peptide resin: 2.8 g of the above peptide resin was added toSwell in 30mL DCM for 30 min. To the above swollen resin was added 150mL of cleavage/deprotection mixture (TFA/H) at room temperature 2 O/EDT/TIS/DCM, 6:3:1.5:3:86.5, v/v) to remove Mmt protection of the Cys side chain for 20 min. The supernatant of the lysis mixture was drained into a flask containing 150mL of water. The deprotection and cleavage procedure was repeated at room temperature for 20min using another 150mL cleavage/deprotection mixture. After removing the solid resin by filtration, the cleavage solutions containing the partially deprotected linear peptide were combined and concentrated under vacuum using a rotary evaporator. The residue (crude peptide) was then lyophilized, yielding 1.2g of partially deprotected crude peptide.
Cyclization: to 1.2g of the above crude peptide solution in 60mL of DMF was added 10mL of MeCN containing 200mg of 1, 2-bis (bromomethyl) benzene. The solution was mixed well and then 890mL MeCN and 240mL water were added. The pH of the reaction mixture was adjusted to pH 8-9 using 1M ammonium carbonate solution. The cyclization reaction was allowed to proceed for 1h at room temperature under magnetic stirring. Cyclization completion was confirmed by MS. The reaction mixture was then divided into three equal portions and freeze-dried in a flask.
Deprotection of the side chain of cyclized peptide: to each lyophilized crude cyclized peptide flask was added 10mL of a solution prepared from TFA/EDT/TIS/H 2 Cleavage mixture of O/Thioanisole/phenol (81.5 mL TFA, 2.5mL EDT, 1.5mL TIS, 5.0mL H per 100mL solution) 2 O, 5.0mL thioanisole, and 5.0g phenol). The cleavage reaction was allowed to proceed at 30 ℃ for 60 min. To the cleavage mixture was added 4 volumes of cold diethyl ether. The crude peptide precipitate was isolated by centrifugation at 3000rpm for 2 min. The crude peptide precipitate was washed three times with cold ether. The crude peptide was dried under vacuum and then subjected to lysine side chain modification according to the following procedure without further purification.
Modification of lysine side chains using acetone-D6: 125mg of the above crude product was dissolved in 12.5mL of a solution consisting of acetic acid, acetone-D6, ethanol (2:2:8.5, v/v), and 324mg of sodium cyanoborohydride (NaBH) 3 CN). The reductive amination reaction was carried out at 30 ℃ for 2 h. The reaction mixture was diluted with water and loaded onto a preparative HPLC column. Purification of isotopically labeled cyclic peptides to purity>90% and freeze-driedAnd (5) drying.
Preparation of PDC-CXCR4 peptide-paclitaxel drug conjugate
Figure BDA0003765213480000281
452mg of 2' -bromoacetyl paclitaxel prepared above and 905mg of cyclized CXCR4 peptide were dissolved in 100mL of MeCN/water (1:1, v/v). The solution is adjusted to pH 7-7.5 using 0.5M ammonium bicarbonate. At room temperature, MS confirmed that the conjugation reaction was complete in about half an hour. The final product was purified using a reversed phase preparative column Daisogel (50X 250mm, 8 μm); mobile phase-solvent a: 0.1% TFA water; and (3) solvent B: 0.1% TFA acetonitrile. Fractions containing the desired product MB1707-D12 were combined and lyophilized (TFA salt). Salt exchange as described above gives the peptide in acetate. The final peptide product was 99.01% pure by HPLC; MW calibration: 2626.20 Da; observed MW value is 2623.35 Da.
Example 3 Cyclo [ Ac-HCys-Tyr-Lys (iPr) -d-Arg-2Nal-Gly-Cys]-Lys(iPr)-PEG6- Synthesis of COOH (a disulfide ring hCys1-Cys7)
Figure BDA0003765213480000291
The sequence hCys (Trt) -Tyr (tBu) -Lys (iPr, Boc) - (D-Arg (Pbf) -2Nal-Gly-Cys (Trt) -Lys (iPr, Boc) -PEG6(SEQ ID NO: 11) was assembled by standard Fmoc chemistry using 2-chlorotriacylchloride resin. Briefly, 4.0 g of resin was swollen in DCM for 2h, washed four times with DMF and then once with DCM. The loading of the first residue Fmoc-PEG6 was performed in DCM using 4 equivalents of amino acid activated with five equivalents of DIEA. After 1.5h of coupling at room temperature, unreacted substitution sites were blocked with methanol/DIEA (1:1, v/v, 24mL) for 30 min. The Fmoc protection was removed in DMF containing 20% piperidine for 20min at room temperature and washed several times with DMF. Ninhydrin test was negative. Stepwise chain assembly of linear peptides was continued by deprotection, activation and coupling cycles using standard Fmoc chemistry.
After coupling the last residue Fmoc-hcys (trt) -OH, Fmoc protection was again removed for 20min using 20% piperidine in DMF. N-terminal acetylation was performed with 5mL of a mixture of acetic anhydride/DIEA/DMF (1:1:4, v/v/v) at room temperature for 30 min. The resin was then washed three times with DMF and then twice with DCM and dried under vacuum.
At room temperature, per gram of crude peptide resin (81.5 mL of EDT, 2.5mL of EDT, 1.0mL of IS, 5.0mL of H per 100mL of solution) 2 O, 5.0mL thioanisole and 5.0 grams phenol) with 10mL TFA/EDT/TIS/H 2 The O/thioanisole/phenol cleavage mixture was subjected to crude peptide cleavage and side chain protection for 70 min. Eight volumes of methyl tert-butyl ether were then added to the cleavage mixture. The crude peptide precipitate was separated by centrifugation at 3000rpm for 3 min. The crude peptide precipitate was washed three times with methyl tert-butyl ether. The crude peptide was then dissolved in water acetonitrile and lyophilized.
The crude lyophilized product was used directly for the cyclization reaction. The lyophilized crude peptide was dissolved in 20% acetic acid at 0.5 mg/mL. Under stirring, a 0.03% mol/L iodine solution was added until the peptide solution became light yellow. The solution is protected from visible light during the cyclization. Cyclization was complete within 0.5h as monitored by mass spectrometry. The cyclized final product was purified using a reverse phase preparative column, Daisogel (50 × 250mm, 8 μm), mobile phase a: 0.1% TFA water; b: 0.1% TFA acetonitrile. Fractions containing the desired product were combined and lyophilized (TFA salt).
Example 4.Cyclo [ Ac-hCys-Tyr-Lys (iPr) -d-Arg-2Nal-Gly-Cys]-Lys(iPr)-PEG6-Lys (a disulfide ring hCys1-Cys7) Synthesis
Figure BDA0003765213480000301
The sequence hCys (Trt) -Tyr (tBu) -Lys (iPr, Boc) - (D-Arg (Pbf) -2Nal-Gly-Cys (Trt) -Lys (iPr, Boc) -PEG6-Lys (Boc)) (SEQ ID NO: 13) was synthesized by standard Fmoc chemistry using RinkAM resin. Briefly, 3.6 g of rink am resin was swollen in DCM for 14h and then washed four times with DMF. Fmoc was removed with 20% piperidine in DMF for 20min at room temperature and washed several times with DMF. Ninhydrin test was negative. Chain assembly was performed stepwise starting from the C-terminus of the linear peptide and completed in nine major steps. In step 1, three equivalents of the protected amino acid Fmoc-Lys (Boc) were activated with DIC/HOBt containing DMF and coupled to Fmoc-depleted RinkAM resin described above for 2h at room temperature followed by Fmoc removal with 20% piperidine in DMF for 20 min. In step 2, three equivalents of Fmoc-PEG6 acid were activated with DCC/HOBt containing DMF and coupled to the deprotected resin in step 1. The appropriate procedure was continued using Fmoc protected amino acids, respectively, until Fmoc hCys (Trt) -OH coupling.
After coupling the last residue Fmoc-hcys (trt) -OH, Fmoc protection was removed again for 20min using DMF with 20% piperidine. N-terminal acetylation was performed with 5mL of a mixture of acetic anhydride/DIEA/DMF (1:1:4, v/v/v) at room temperature for 30 min. The resin was then washed three times with DMF, then twice with DCM and dried under vacuum.
From the resin at room temperature with TFA/EDT/TIS/H 2 O/Thioanisole/phenol cleavage mixture (81.5 mL of EDT, 2.5mL of EDT, 1.0mL of IS, 5.0mL of H per 100mL of solution) 2 O, 5.0mL thioanisole, and 5.0g phenol) and cleaving the final peptide for 70 min. Eight volumes of methyl tert-butyl ether were added to the cleavage mixture. The crude peptide precipitate was separated by centrifugation at 3000rpm for 3 min. The crude peptide precipitate was washed three times with methyl tert-butyl ether. The crude peptide was then dissolved in aqueous acetonitrile and lyophilized.
The crude lyophilized product was used directly for the cyclization reaction. The lyophilized crude peptide was dissolved at a rate of 0.5mg/mL in water containing 20% acetic acid (500 mg crude peptide per liter). To the peptide solution was added 0.03% mol/L iodine solution with stirring until the solution became pale yellow. The cyclization was completed in 0.5h in the dark by mass spectrometric monitoring. The cyclized final product was purified using a reverse phase preparative column Daisogel (50X 250mm, 8 μm); mobile phase-solvent a: 0.1% TFA water; solvent B: 0.1% TFA acetonitrile. Fractions containing the desired product were combined and lyophilized (TFA salt).
Example 5.Cyclo [ Acetyl-hCys-Tyr-Lys (iPr) -d-Arg-2Nal-Gly-Cys]-Lys(iPr)- mini-PEG 6-Cys-amide, a disulfide ring, hCys1-Cys 7.
Figure BDA0003765213480000321
The sequence hCys (Mmt) -Tyr (tBu) -Lys (iPr, Boc) - (D-Arg (Pbf) -2Nal-Gly-Cys (Mmt) -Lys (iPr, Boc) -PEG6-Cys (Trt) (SEQ ID NO: 15) was assembled by standard Fmoc chemistry using Sieber amide AM resin (Xi' an LanXiao Chemical Limited, Sichuan). Briefly, 4g of Sieber amide AM resin was swollen in 30mL of DCM for 2h, then washed four times with DMF. Fmoc was removed in 25mL of 20% piperidine in DMF at room temperature for 20min and washed several times with DMF. Ninhydrin test was negative. Chain assembly was performed stepwise starting from Fmoc-Cys (Trt) -OH at the C-terminus of the linear peptide. Three equivalent protected amino acids Fmoc-Cys (Trt) -OH were activated with 8mL of DIC/HOBt containing DMF and coupled with Fmoc-depleted Sieber amide AM resin prepared above for 2h at room temperature. Ninhydrin test was negative. Capping with 16mL of a mixture of acetic anhydride/DIEA/DCM at a volume ratio of 1:1:4 for 30min of unreacted amino group. Subsequently, Fmoc was removed with 25mL of 20% piperidine in DMF for 20 min. The following residues were coupled sequentially without capping: fmoc- (mini-PEG6) -OH, Fmoc-Lys (iPr, Boc) -OH, Fmoc-Cys (Mmt) -OH, Fmoc-Gly-OH, Fmoc-2Nal-OH, Fmoc-D-Arg (Pbf) -OH, Fmoc-Lys (iPr, Boc) -OH, moc-Tyr (tBu) -OH and Fmoc-hCys (Mmt) -OH. After coupling the final residue Fmoc hCys (Mmt), Fmoc protection was removed again for 20min using 25mL DMF with 20% piperidine. Acetylation of the N-terminus was performed at room temperature for 30min using 16mL of a mixture of acetic anhydride/DIEA/DMF (1:1:4, v/v/v). The resin was then washed three times with 25mL DMF followed by three times with 25mL DCM, and then dried under vacuum to give 8 g of peptide resin containing the assembled target linear peptide. The dried resin was used in the following operations.
Removal of the Cys residue protection by Mmt and cleavage of the partially protected linear peptide from the peptide resin:about 2 grams of the above peptide resin was swollen in 30mL DCM for 30 min. To the swollen resin above was added 100mL of cleavage/deprotection mixture (TFA/H) 2 O/EDT/TIS/DCM, 6:3:1.5:3:86.5, v/v) to remove Mmt protection of the Cys side chain. The mixture was stirred at room temperature for 20 min. At room temperatureThe deprotection and cleavage procedure was repeated with another 100mL cleavage/deprotection mixture for 20 min. After removing the solid resin by filtration, the cleavage solutions containing the partially deprotected linear peptide were combined and concentrated under vacuum using a rotary evaporator. The residue (crude peptide) was then lyophilized to give about 1 g of partially deprotected crude peptide.
The crude lyophilized product was used directly for the cyclization reaction. The lyophilized crude peptide was dissolved at 0.5mg/mL in water containing 20% acetic acid (500 mg crude peptide per liter). To the peptide solution was added 0.03% mol/L iodine solution with stirring until the solution became light yellow. Cyclization was completed in 0.5h in the dark, monitored by mass spectrometry. The cyclization reaction was carried out at room temperature for 1 h. The reaction mixture was then divided into several equal portions and freeze-dried.
Deprotection of the side chain of cyclized peptide: cleavage cocktail (TFA/EDT/TIS/H) was used 2 O/Thioanisole/phenol containing 81.5mL of EDT, 2.5mL of EDT, 1.5mL of IS, 5.0mL of H per 100mL of solution 2 O, 5.0mL thioanisole and 5.0g phenol) was performed on the lyophilized crude cyclized peptide for 60min, 10mL in each vessel. To the cleavage mixture was added 4 volumes of cold diethyl ether. The crude peptide precipitate was separated by centrifugation at 3000rpm for 2 min. The crude peptide precipitate was washed three times with diethyl ether. The crude peptide was purified on preparative HPLC to purity>90% and freeze-drying.
Example 6 preparation of PDC-CXCR4 peptide conjugate Compound F
Figure BDA0003765213480000341
450mg of 2' -bromoacetyl paclitaxel prepared above and 905mg of cyclized CXCR4 peptide Compound C were dissolved in 100mL of MeCN/water (1:1, v/v). The solution is adjusted to pH 7-7.5 using 0.5M ammonium bicarbonate. The coupling reaction was monitored by mass spectrometry and the reaction was complete in about half an hour. The final product was purified using a reverse phase preparative column Daisogel (50X 250mm, 8 μm); mobile phase-solvent a: 0.1% TFA water; and (3) solvent B: 0.1% TFA acetonitrile. Fractions containing the desired product compound F were combined and lyophilized (TFA salt). Salt exchange as described above gives the peptide in the form of acetate.
Example 7 preparation of Compounds G-P
Compound G-P was prepared in a similar manner, starting from Rink amide resin or 2-chlorotriphenyl resin or Sieber amide AM resin, using standard Fmoc solid phase peptide chemistry.
Figure BDA0003765213480000351
Figure BDA0003765213480000361
Figure BDA0003765213480000371
Figure BDA0003765213480000381
Figure BDA0003765213480000391
EXAMPLE 8 Synthesis of isotopically labeled Compounds
Deuterated or tritiated cyclic peptide moieties are prepared using the methods described in the above examples, and using the corresponding deuterated compound B-D12 or the corresponding tritiated compound (i.e., compound B-T12, for tritiated B compounds, analogous to B-D12, but where deuterium is replaced with tritium). Using these deuterium-labeled (B-D12) and tritium-labeled (B-T12) compounds, the corresponding deuterium-labeled ("-D12") and tritium-labeled (i.e., "-T12") compounds A-P, i.e., compounds A-D12, A-T12, B-D12, B-T12, C-D12, C-T12, … P-D12, and P-T12, can be synthesized.
125 Human CXCR4/I-SDF-1 alpha binding inhibition assay: (by EUROFINS CEREP SA, Le Bois l 'Ev request, 86600Celle l' Evescault, france, performance): human chemokine receptor CXCR4 expressed in Chem-1 cells was used in modified HEPES buffer pH 7.4. An aliquot of 0.5. mu.g (membrane protein may vary from batch to batch, if necessary, the concentration used will be adjusted) is incubated with 0.03nM [ 2], [ 2] 125 I]SDF-1. alpha. was incubated for 90 min. Nonspecific binding was estimated in the presence of 30nM SDF-1 α. Filtering and washing the membrane, and then counting the filtrate to determine [ 2], [ 125 I]SDF-1 alpha binds specifically. Starting from 10. mu.M, compounds were screened by serial dilution with 11 spots (Vallenzuela Fernandez A et al, J Biol chem.277(18):15677,2002)). The antagonist peptides and drug conjugates thereof disclosed herein have a CXCR4 binding affinity of 1.0 μ M or less for practical therapeutic use.
The foregoing discussion of the invention has been presented for purposes of illustration and description. The foregoing is not intended to limit the invention to the form or forms disclosed herein. Although the description of the invention includes description of one or more embodiments and certain variations and modifications, other variations and modifications are within the scope of the invention, e.g., as may be within the skill and knowledge of those in the art, after understanding the present invention. The invention is intended to obtain rights which include alternative embodiments to the extent permitted, including alternate, interchangeable and/or equivalent structures, functions, ranges or steps to be claimed, whether or not such alternate, interchangeable and/or equivalent structures, functions, ranges or steps are disclosed herein, and without intending to publicly dedicate any patentable subject matter. All references cited herein are incorporated by reference in their entirety.
Sequence listing
<110> mainline bioscience, Inc
<120> isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same
<130> MLB-000310PC
<150> 16/752,690
<151> 2020-01-26
<160> 26
<170> PatentIn version 3.5
<210> 1
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compounds of structural formula I
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> together with the sulfur atom attached thereto is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> form a cyclic structure by a sulfur atom and a linker- (CH2-Ar1-CH2) a-, wherein Ar1 is an optionally substituted aryl group, a is 0 or 1
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or
Lys (iPr); each may be the (L) or (D) -isomer
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> together with the sulfur atom attached thereto is cysteine or homocysteine
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> may be 1 to 4 amino acids, each of which may be independently Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), Lys (iPr), and may be independently (L) -or (D) -isomer.
<400> 1
Xaa Tyr Xaa Arg Xaa Gly Xaa Xaa
1 5
<210> 2
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compounds of structural formula IA
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> together with the sulfur atom attached thereto is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> form a cyclic structure by a sulfur atom and a linker- (CH2-Ar1-CH2) a-, wherein Ar1 is an optionally substituted aryl group, and a is 0 or 1
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or Lys (iPr), each of which may be the (L) or (D) -isomer
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> with the sulfur atom attached thereto is cysteine or homocysteine
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> can be 1 to 4 amino acids, each of which can independently be Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr),
Orn (iPr), Lys (iPr), each independently may be the (L) -or (D) -isomer
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> wherein one amino acid is linked to the-L1-Q moiety
<400> 2
Xaa Tyr Xaa Arg Xaa Gly Xaa Xaa
1 5
<210> 3
<211> 14
<212> PRT
<213> Artificial sequence
<220>
<223> Compound of formula 1A
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> together with the sulfur atom attached thereto is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> form a cyclic structure via a sulfur atom and a linker- (CH2-Ar1-CH2) a-, wherein Ar1 is an optionally substituted aryl group and a is 0 or 1
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or Lys (iPr), each of which may be (L) or (D) -isomer
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> together with the sulfur atom attached thereto is cysteine or homocysteine
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> may be 1 to 4 amino acids, each of which may be independently Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), Lys (iPr), and each of which may be independently (L) -or (D) -isomer.
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> L1a is a polymer linker having a functional group to link Q, wherein L1 comprises about 2 to about 20 monomers or copolymers;
<220>
<221> misc_feature
<222> (10)..(10)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> MISC_FEATURE
<222> (11)..(11)
<223> is Y1, a side chain functional group of amino acid AA4
<220>
<221> MISC_FEATURE
<222> (12)..(12)
<223> is L2, a non-polymeric linker
<220>
<221> MISC_FEATURE
<222> (13)..(13)
<223> is a functional group of the functional group Y2 or L2 of the compound having medical value;
<220>
<221> MISC_FEATURE
<222> (14)..(14)
<223> is Z, a compound having a medicinal value
<400> 3
Xaa Tyr Xaa Arg Xaa Gly Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5 10
<210> 4
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound A
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetylation on alpha-amino groups
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> formation of a cyclic structure by a sulfur atom and a linker-CH 2-phenyl-CH 2
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> attachment of thiol-functional cysteine to paclitaxel via a-CH 2-C (= O) -linker
<400> 4
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 5
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Sieber amide AM resin-binding peptide of example 1
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> protection by Fmoc and Mmt
<220>
<221> MISC_FEATURE
<222> (2)..(2)
<223> protected by tBu
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> Pbf protected (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> protected by Mmt
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> mini-PEG6 linker
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> Trt protection and ligation to Sieber amide AM resin
<400> 5
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 6
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound B
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> alpha-amino acetylation
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> formation of a cyclic structure by a sulfur atom and a linker-CH 2-phenyl-CH 2
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 6
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 7
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> peptide chain attached to Sieber amide AM resin in example 2
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> Fmoc and Mmt protection
<220>
<221> MISC_FEATURE
<222> (2)..(2)
<223> protected by tBu
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> protection by Boc
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> Pbf protected (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> protected by Mmt
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> protection by Boc
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> mini-PEG6 linker
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> protected by Trt and attached to Sieber amide AM resin
<400> 7
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 8
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound B-D12 (deuterated)
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> alpha-Aminoacetylation
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> formation of a cyclic structure by a sulfur atom and a linker-CH 2-phenyl-CH 2
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution with deuterated iPr at side chain amino group
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution with deuterated iPr group on side chain amino group
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form an amine bond with Lys and an amide bond with alpha-amino group of cysteine
<400> 8
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 9
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound A-D12 (deuterated Compound A)
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetylation on alpha-amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> formation of a cyclic structure by a sulfur atom and a linker-CH 2-phenyl-CH 2
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution with deuterated iPr group on side chain amino functional group
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2NaI
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution with deuterated iPr group on side chain amino functional group
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form an amine bond with Lys and an amide bond with alpha-amino group of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> attachment of thiol-functional cysteine to paclitaxel via a-CH 2-C (= O) -linker
<400> 9
Cys Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 10
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compound C of example 3
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetylation on alpha-amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution with iPr group on side chain amino functional group
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> iPr radical substitution on side chain amino function group
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution with PEG-6 linker having modified functional group at both ends, formation of amide bond with carboxyl terminal of Lys, and termination with-CH 2-CH2-CO2H group at the other end
<400> 10
Cys Tyr Lys Arg Xaa Gly Cys Lys
1 5
<210> 11
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> 11 hCys protein analogs
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> Trt-protected homocysteine
<220>
<221> MISC_FEATURE
<222> (2)..(2)
<223> tBu protected
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> iPr and Boc substituted side chain amino groups
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> Pbf protected (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> Trt protected
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> amide attached to PEG6 Polymer
<400> 11
Xaa Tyr Lys Arg Xaa Gly Cys Lys
1 5
<210> 12
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound D (protein 12)
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> homocysteine with acetylated substituent on alpha-amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 12
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10
<210> 13
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> homocysteine analogue starting Material
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> Trt-protected homocysteine
<220>
<221> MISC_FEATURE
<222> (2)..(2)
<223> protected by tBu
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> Pbf protected (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> Trt protected
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> protection by Boc
<400> 13
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10
<210> 14
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound E (Peptide 14)
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> homocysteine with acetylated substituent on alpha-amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 14
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 15
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> peptide 15-Linear peptide starting Material
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> homocysteine with Mmt protecting group
<220>
<221> MISC_FEATURE
<222> (2)..(2)
<223> protected by tBu group
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> Pbf protecting group-bearing (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (7)..(7)
<223> protected by Mmt
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr and Boc
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> PEG6 Polymer linker
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> Trt protected
<400> 15
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 16
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound F
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> homocysteine with acetylated substituent on alpha-amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between homocysteine and cysteine group sulfur atom
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> thiol functional group of cysteine to paclitaxel via-CH 2-C (= O) -linker
<400> 16
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 17
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compound G-attachment to eribulin
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetyl protected homocysteine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> the terminus of Lys is linked to eribulin via a PEG6 linker and a-CH 2-C (= O) -linker
<400> 17
Xaa Tyr Lys Arg Xaa Gly Cys Lys
1 5
<210> 18
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound H comprising paclitaxel
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> Ring Structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> thiol functional group of cysteine to paclitaxel via-CH 2-C (= O) -linker
<400> 18
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 19
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound I-attachment to paclitaxel
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetylated homocysteine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> formation of a cyclic structure by a sulfur atom and a-CH 2-phenyl-CH 2-linker
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form an amine bond with Lys and an amide bond with alpha-amino group of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> thiol functional group of cysteine to paclitaxel via-CH 2-C (= O) -linker
<400> 19
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 20
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compound J-ligation to eribulin
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> the carboxyl terminal of Lys is linked to a PEG7 polymer linker with modified functional groups at both ends to form an amide bond with Lys, and the other end of PEG7 is linked to eribulin through an amide bond.
<400> 20
Xaa Tyr Lys Arg Xaa Gly Cys Lys
1 5
<210> 21
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> Compound K-CXCR4 peptide linker
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> Ring Structure formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> the side chain amino is replaced by iPr, and is replaced by a functional group PEG-6 linker with modified two ends, an amido bond is formed with the carboxyl terminal of Lys, and the other end is terminated by-CH 2-CH2-CO2H group
<400> 21
Xaa Tyr Lys Arg Xaa Gly Cys Lys
1 5
<210> 22
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound L-CXCR4 linker capped with Cys
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 22
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Cys
1 5 10
<210> 23
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound M
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 23
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10
<210> 24
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound N
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> acetylated homocysteine
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> substitution of side chain amino group by azide
<400> 24
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10
<210> 25
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound O-is analogous to Compound N containing 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> 3-mercaptopropionic acid
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<220>
<221> MISC_FEATURE
<222> (10)..(10)
<223> substitution of side chain amino group by azide
<400> 25
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10
<210> 26
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<223> Compound P
<220>
<221> MISC_FEATURE
<222> (1)..(1)
<223> homocysteine with acetylated substituent on alpha amino group
<220>
<221> MISC_FEATURE
<222> (1)..(7)
<223> the cyclic structure is formed by disulfide bond between sulfur atoms of two groups
<220>
<221> MISC_FEATURE
<222> (3)..(3)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (4)..(4)
<223> (D) -isomer
<220>
<221> MISC_FEATURE
<222> (5)..(5)
<223> 2Nal
<220>
<221> MISC_FEATURE
<222> (8)..(8)
<223> substitution of side chain amino group by iPr
<220>
<221> MISC_FEATURE
<222> (9)..(9)
<223> a PEG-7 linker having modified functional groups at both ends to form amine bonds with Lys and amide bonds with alpha-amino groups of cysteine
<400> 26
Xaa Tyr Lys Arg Xaa Gly Cys Lys Xaa Lys
1 5 10

Claims (33)

1. An isotopically-labeled selective CXCR4 binding peptide conjugate ("PC") of the formula:
Figure FDA0003765213470000011
or a pharmaceutically acceptable salt thereof,
wherein:
a is 0 or 1;
b is an integer between 1 and 4;
AA 1 and the sulfur atom to which it is attached is 3-mercaptopropionic acid, optionally substituted cysteine, optionally substituted homocysteine, or optionally substituted penicillamine;
AA 2 and the sulfur atom to which it is attached is cysteine or homocysteine;
Ar 1 is an optionally substituted aryl group;
X 1 is Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), or Lys (iPr);
AA 3 each is independently Gly, Phe, 2Nal, 1Nal, Arg, Dap, Dab, Orn, Lys, Dap (iPr), Dab (iPr), Orn (iPr), Lys (iPr);
and wherein AA 1 、X 1 、AA 2 Or AA 3 Comprises the following structural moiety:
-L 1 –Q,
wherein
L 1 Is a polymer linker having a functional group for linking Q, wherein L 1 Comprising from about 2 to about 20 monomers or copolymers;
q is absent, is AA 4 Or a structural formula: - [ AA 4 -Y 1 -L 2 ] c -Y 2 A part of-Z and a part of-Z,
wherein
c is 0 or 1;
AA 4 is an amino acid or a derivative thereof;
Y 1 is amino acid AA 4 A side chain functional group of (1);
L 2 is a non-polymeric linker;
z is a compound of pharmaceutical value; and is
Y 2 Is a functional group or L of the compound having a medicinal value 2 A functional group of (a);
wherein, AA 1 、AA 2 、X 1 、AA 3 And AA 4 With one or more of a positron-emitting radioisotope, a radioactive metal isotope, deuterium, tritium, a, 13 C、 14 C、 18 O or a combination thereof.
2. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein the compound of pharmaceutical value comprises an imaging agent, a contrast agent or a therapeutic agent.
3. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1 having the structural formula:
Figure FDA0003765213470000021
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、L 1 Q and Ar 1 As defined in claim 1.
4. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 3, wherein Q is deleted.
5. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 3, wherein Q is AA 4 Or the structural formula: - [ AA 4 –Y 1 –L 2 ] c –Y 2 A moiety of-Z, wherein c, AA 4 、Y 1 、L 2 、Y 2 And Z is as defined in claim 3.
6. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein a is 0.
7. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein a is 0.
8. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 1 Is homocysteine.
9. The isotopically labeled selective CXCR4 binding peptide conjugate of claim, wherein a is 1.
10. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein X is 1 Is labeled with an isotope.
11. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 3 Is labeled with an isotope.
12. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein X is 1 And AA 3 Is isotopically labeled, and wherein the isotopic labeling comprises deuterium, tritium, deuterium, tritium, deuterium, hydrogen, oxygen, hydrogen, and mixtures thereof, 13 C、 14 C、 18 O or a combination thereof.
13. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 10, wherein X is 1 And AA 3 Is labelled with deuterium or tritium isotopes.
14. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein the compound of pharmaceutical value is a therapeutic agent.
15. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 12, wherein the therapeutic agent is an anti-cancer agent.
16. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 13, wherein the anti-cancer agent is selected from the group consisting of paclitaxel, altretamine, asparaginase, bleomycin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, diethylstilbestrol, eribulin, ethinylestradiol, etoposide, mitomycin, o-chlorobenzenedichloroethane, mitoxantrone, pentostatin, bromopropylpiperazine, mithramycin, prednisone, procarbazine, streptozotocin, tamoxifen, teniposide, vinblastine and vincristine.
17. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1 having the structural formula:
Figure FDA0003765213470000031
wherein a, b, AA 1 、X 1 、AA 2 、AA 3 、AA 4 、Y 1 、Y 2 、L 2 Z and Ar 1 As defined in claim 1; l is a radical of an alcohol 1a Is a polymer linker comprising from about 2 to about 20 monomers or copolymers.
18. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein L is 2 Is of the formula-CH 2 –C(=O)-NH–CH 2 –、–CH 2 –NH–C(=O)–CH 2 –、(CH 2 ) n -C(=O)–NH–CH 2 –、(CH 2 ) n -NH-C(=O)–CH 2 -, or- (CH) 2 ) n The non-polymeric linker of (a), wherein n is an integer from 1 to 6.
19. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1,wherein AA 1 And the sulfur atom attached thereto is an optionally substituted cysteine.
20. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 17, wherein AA is 1 Is substituted by acetyl.
21. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 2 And the sulfur atom attached thereto is an optionally substituted cysteine.
22. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein X is 1 Is Lys (iPr) or Lys (deuterated iPr).
23. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 3 Is Lys (iPr) or Lys (deuterated iPr).
24. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein Ar is 1 Is phenyl.
25. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein b is 1.
26. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 23, wherein AA 3 Is Lys (iPr) or Lys (deuterated iPr).
27. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 24, wherein L is 1 Is a polymeric linker of the formula: -NH- (CH) 2 ) 2 –[O–CH 2 –CH 2 –]n–O-(CH 2 ) 2 -C (═ O) -, where n is an integer from 2 to 20, -NH-linking amino acid AA 3 Of (a) carbonylA radical carbon, and-C (═ O) -linking amino acids AA 4 The alpha-amino functional group of (a).
28. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 4 Is cysteine, homocysteine or penicillin amine.
29. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein AA is 1 、X 1 、AA 2 Or AA 3 At least one of the amino acid residues is the (D) -isomer.
30. The isotopically labeled selective CXCR4 binding peptide conjugate of claim 1, wherein the selective CXCR4 binding peptide conjugate is selected from the group consisting of: the compound of the structural formula A, the compound of the structural formula B, the compound of the structural formula C, the compound of the structural formula D, the compound of the structural formula E, the compound of the structural formula F, the compound of the structural formula G, the compound of the structural formula H, the compound of the structural formula I, the compound of the structural formula J, the compound of the structural formula K, the compound of the structural formula L, the compound of the structural formula M, the compound of the structural formula N, the compound of the structural formula O, the compound of the structural formula P, corresponding deuterium labeled compounds and corresponding tritium labeled compounds.
31. A method of treating a subject having cancer, the method comprising administering to the subject a therapeutically effective amount of an isotopically-labeled selective CXCR4 binding peptide conjugate of claim 1, wherein the compound of pharmaceutical value is an anti-cancer agent.
32. The method of claim 29, wherein the anti-cancer agent is selected from the group consisting of: paclitaxel, altretamine, asparaginase, bleomycin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, diethylstilbestrol, eribulin, ethinylestradiol, etoposide, mitomycin, o-chlorobenzenedichloroethane, mitoxantrone, pentostatin, bromopropylpiperazine, mithramycin, prednisone, procarbazine, streptozotocin, tamoxifen, teniposide, vinblastine, and vincristine.
33. The method of claim 29, wherein the cancer is selected from the group consisting of breast cancer, lung cancer, pancreatic cancer, renal cancer, brain cancer, blood cancer, leukemia, prostate cancer, ovarian cancer, and bladder cancer.
CN202080094623.3A 2020-01-26 2020-03-27 Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same Pending CN115003684A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16/752,690 US11123437B2 (en) 2017-09-05 2020-01-26 Selective CXCR4 binding peptide conjugate and methods for making and using the same
US16/752,690 2020-01-26
PCT/US2020/025522 WO2021150258A1 (en) 2020-01-26 2020-03-27 Isotopically labelled selective cxcr4 binding peptide conjugate and methods for making and using the same

Publications (1)

Publication Number Publication Date
CN115003684A true CN115003684A (en) 2022-09-02

Family

ID=76992501

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080094623.3A Pending CN115003684A (en) 2020-01-26 2020-03-27 Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same

Country Status (4)

Country Link
EP (1) EP4097120A1 (en)
JP (1) JP2023516878A (en)
CN (1) CN115003684A (en)
WO (1) WO2021150258A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101678213A (en) * 2007-05-30 2010-03-24 伊莱利利公司 Cyclic peptide cxcr4 antagonists
US20110027175A1 (en) * 2006-02-27 2011-02-03 Technische Universitat Munchen Cancer imaging and treatment
CN102626522A (en) * 2012-04-12 2012-08-08 韩彦江 Polypeptide radioactive diagnosis and treatment medicament based on chemotactic factor receptor CXCR4 polypeptide antagonist
CN102869675A (en) * 2010-01-26 2013-01-09 国家研究委员会 Cyclic peptides binding CXCR4 receptor and relative medical and diagnostic uses
US20130156723A1 (en) * 2009-10-29 2013-06-20 Lucia Irene Gonzalez Synthetic stereoisomer peptides in the retro-inverso and inverso configuration, and with cyclic and linear structure, their polymer conjugates, their encapsulation in polymer particles, and uses thereof
AU2012381808A1 (en) * 2012-06-06 2015-01-22 Polyphor Ag Beta-hairpin peptidomimetics
AU2014396468A1 (en) * 2014-06-06 2016-12-01 Technische Universitat Munchen Modified cyclopentapeptides and uses thereof
WO2019050564A1 (en) * 2017-09-05 2019-03-14 Mainline Biosciences High affinity cxcr4 selective binding conjugate and method for using the same
CN109922818A (en) * 2016-09-06 2019-06-21 主线生物科学公司 CXCR4 antagonist and application method

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR122020024394B1 (en) * 2010-12-29 2021-05-11 F. Hoffmann-La Roche Ag conjugate and pharmaceutical composition

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110027175A1 (en) * 2006-02-27 2011-02-03 Technische Universitat Munchen Cancer imaging and treatment
CN101678213A (en) * 2007-05-30 2010-03-24 伊莱利利公司 Cyclic peptide cxcr4 antagonists
US20130156723A1 (en) * 2009-10-29 2013-06-20 Lucia Irene Gonzalez Synthetic stereoisomer peptides in the retro-inverso and inverso configuration, and with cyclic and linear structure, their polymer conjugates, their encapsulation in polymer particles, and uses thereof
CN102869675A (en) * 2010-01-26 2013-01-09 国家研究委员会 Cyclic peptides binding CXCR4 receptor and relative medical and diagnostic uses
CN102626522A (en) * 2012-04-12 2012-08-08 韩彦江 Polypeptide radioactive diagnosis and treatment medicament based on chemotactic factor receptor CXCR4 polypeptide antagonist
AU2012381808A1 (en) * 2012-06-06 2015-01-22 Polyphor Ag Beta-hairpin peptidomimetics
AU2014396468A1 (en) * 2014-06-06 2016-12-01 Technische Universitat Munchen Modified cyclopentapeptides and uses thereof
CN109922818A (en) * 2016-09-06 2019-06-21 主线生物科学公司 CXCR4 antagonist and application method
WO2019050564A1 (en) * 2017-09-05 2019-03-14 Mainline Biosciences High affinity cxcr4 selective binding conjugate and method for using the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SHENG-BIN PENG等: ""Identification of LY2510924, a Novel Cyclic Peptide CXCR4 Antagonist That Exhibits Antitumor Activities in Solid Tumor and Breast Cancer Metastatic Models"", 《MOLECULAR CANCER THERAPEUTICS》 *

Also Published As

Publication number Publication date
EP4097120A1 (en) 2022-12-07
WO2021150258A1 (en) 2021-07-29
WO2021150258A9 (en) 2021-11-11
JP2023516878A (en) 2023-04-21

Similar Documents

Publication Publication Date Title
KR100625708B1 (en) Peptides and Compounds That Bind to a Receptor
KR101169846B1 (en) Cyclic peptide cxcr4 antagonists
US7521419B2 (en) Peptide-based compounds
CN111183146A (en) Conjugates that bind CXCR4 with high affinity selectivity and methods of use thereof
CN104039342A (en) Peptidomimetic macrocyles
US11883501B2 (en) Selective CXCR4 binding peptide conjugate and methods for making and using the same
US10870681B2 (en) CXCR4 antagonists and methods of use
CN115003684A (en) Isotopically labeled selective CXCR4 binding peptide conjugates and methods of making and using same
US10639379B2 (en) High affinity CXCR4 selective binding conjugate and method for using the same
AU2003253258A1 (en) Fluoro-alkyl-cyclopeptide derivatives having anti-integrin activity
NL2025303B1 (en) Coronavirus targeting drugs
EP4048324A1 (en) Fibrin-binding compounds for imaging and treatment

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20220914

Address after: Building 2, No. 1666, Xinyang Road, Lingang New Area, Pudong New Area, Shanghai

Applicant after: Mainstream Biotechnology (Shanghai) Co.,Ltd.

Address before: Pennsylvania, America

Applicant before: Mainline Biosciences Ltd.

TA01 Transfer of patent application right