CN117396234A - Compositions and methods for treating prostate cancer - Google Patents

Compositions and methods for treating prostate cancer Download PDF

Info

Publication number
CN117396234A
CN117396234A CN202280037818.3A CN202280037818A CN117396234A CN 117396234 A CN117396234 A CN 117396234A CN 202280037818 A CN202280037818 A CN 202280037818A CN 117396234 A CN117396234 A CN 117396234A
Authority
CN
China
Prior art keywords
pharmaceutical composition
antibody
μci
seq
radiometal
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
CN202280037818.3A
Other languages
Chinese (zh)
Inventor
R·萨尔特
N·萨克塞纳
C·迪维吉
J·奥多诺休
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.)
Janssen Biotech Inc
Original Assignee
Janssen Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Biotech Inc filed Critical Janssen Biotech Inc
Priority claimed from PCT/IB2022/054891 external-priority patent/WO2022249089A1/en
Publication of CN117396234A publication Critical patent/CN117396234A/en
Pending legal-status Critical Current

Links

Landscapes

  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Embodiments of the present invention provide compositions and methods for treating cancer, particularly prostate cancer. According to certain embodiments, a method of treating cancer in a patient comprises administering to the patient a therapeutically effective amount of a radioconjugate, wherein the radioconjugate comprises an antibody or antigen binding domain having binding specificity for hK 2. Also provided herein are pharmaceutical compositions comprising radiolabeled antibodies having binding specificity for hK 2.

Description

Compositions and methods for treating prostate cancer
Cross Reference to Related Applications
The present application claims priority from U.S. provisional patent application 63/193,704 filed on day 5 and 27 of 2021 and U.S. provisional patent application 63/335,761 filed on day 4 and 28 of 2022, both of which are incorporated herein by reference in their entireties for all purposes.
Technical Field
Embodiments of the present invention relate to compositions and methods for treating prostate cancer. In particular, embodiments of the present invention relate to a radioconjugate composition for use in hK 2-targeted therapies.
Electronically submitted reference sequence listing
The present application contains a sequence listing submitted electronically via the EFS-Web as an ASCII format, file name "JBI6423 WORTV1_SeqListing. Txt" and creation date of 2022, 5 month 12 days, and size of 17kb. This sequence listing submitted via EFS-Web is part of this specification and is incorporated by reference herein in its entirety.
Background
Prostate cancer is one of the most common forms of cancer. Tumor growth is typically a process that occurs over a long period of time. Prostate cancer is often a mild form of cancer. In fact, most people diagnosed with prostate cancer survive and recover. A minority of people experience a more aggressive form of prostate cancer, which metastasizes at an early stage. This invasive form of prostate cancer can only heal if diagnosed at an early stage before the cancer has spread to extracapsular tissue.
Patient management patterns for metastatic castration-resistant prostate cancer (mCRPC) have changed as a result of the successor of multiple new agents, including Androgen Receptor (AR) directed therapies (e.g., enzalutamide and abiraterone acetate Long Jiapo), chemotherapy (e.g., docetaxel and cabazitaxel), and cellular immunotherapy (e.g., sipuleucel-T). With these agents, overall survival has improved from the previously reported range of 6 months to 10 months to 18 months to 24 months. However, in prostate cancer patients receiving anti-androgens or androgen synthesis inhibitors, most progress within 13 months to 20 months.
There remains a need for new therapeutic agents and methods for the treatment and diagnosis of prostate cancer; in particular, therapies with mechanisms of action that overcome the resistance pathway are critical in developing alternative strategies for treating mCRPC.
Disclosure of Invention
The present invention relates to pharmaceutical compositions, methods of preparing pharmaceutical compositions, and methods of treating cancer in patients in need of such treatment.
According to one embodiment of the invention, a method of treating cancer in a patient comprises: administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein: the radioconjugate comprises at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK 2; the radioactive metal complex comprises a radioactive metal; and upon administration, the radiometal provides targeted radioactivity of about 50 μci to about 350 μci per dose of the pharmaceutical composition.
According to one embodiment of the invention, a method of treating cancer in a patient comprises: administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein: the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2; the radioactive metal complex comprises 225 A radioactive metal of Ac; and upon administration, the radiometal provides targeted radioactivity of about 50 μci to about 350 μci per dose of the pharmaceutical composition.
According to one embodiment, the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK2, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequences of SEQ ID NO:1 and SEQ ID NO:2 and SEQ ID NO:3 and a light chain variable region comprising the amino acid sequences of SEQ ID NO:4 and SEQ ID NO:5 and SEQ ID NO: 6.
According to one embodiment, the radioactive metal complex comprises a chelator that is DOTA.
According to one embodiment, the radioactive conjugate comprises a radioactive metal chelated to: (a) A compound of formula (IV)
Or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and
R 4 is an antibody; or alternatively
(b) A compound of formula (V)
Or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and
R 4 is an antibody;
for example, wherein the chelating agent is a compound of the formula:
according to one embodiment, the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 25 μci to about 350 μci per about 2mg of total antibody, or about 50 μci to about 350 μci per about 2mg of total antibody.
According to one embodiment, the method comprises administering the pharmaceutical composition intravenously to a patient.
Embodiments of the invention are particularly useful in treating patients who have been diagnosed with prostate cancer; for example, patients with advanced prostate cancer. According to one embodiment, the cancer is non-restricted prostate cancer. According to another embodiment, the cancer is metastatic prostate cancer. According to another embodiment, the cancer is Castration Resistant Prostate Cancer (CRPC). According to another embodiment, the cancer is metastatic castration-resistant prostate cancer (mCRPC). According to another embodiment, the cancer is mCRPC with an adenocarcinoma.
Another embodiment of the present invention provides a pharmaceutical composition comprising a radioconjugate and one or more pharmaceutically acceptable excipients, wherein: the radioconjugate comprises at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2, and the radiometal complex comprises a radiometal.
According to one embodiment, a pharmaceutical composition comprises a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein: the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK2, the radiometal complex comprising 225 Ac, and the antibody comprises a heavy chain variable region comprising the amino acid sequences of SEQ ID NO. 1 and SEQ ID NO. 2 and SEQ ID NO. 3,the light chain variable region comprises the amino acid sequences of SEQ ID NO. 4 and SEQ ID NO. 5 and SEQ ID NO. 6.
According to one embodiment, the one or more pharmaceutically acceptable excipients comprise one or more radioprotectants, such as sodium ascorbate, gentisic acid, or a combination thereof (e.g., in an amount of about 0.1w/v% to about 5w/v%, or about 0.1w/v% to about 4w/v%, or about 0.1w/v% to about 3w/v%, about 0.1w/v% to about 2w/v%, or about 0.1w/v% to about 1w/v%, or about 0.25w/v% to about 0.75w/v%, or about 0.5 w/v%).
According to one embodiment, the one or more pharmaceutically acceptable excipients comprise one or more surfactants, such as polysorbate 20.
According to one embodiment, the pharmaceutical composition comprises a radioactive conjugate, sodium ascorbate, polysorbate 20, an acetate buffer, and water.
According to one embodiment, the pharmaceutical composition comprises an aqueous solution of the radioactive conjugate, about 24mM to 28mM acetate, about 0.25w/v% to 0.75w/v% sodium ascorbate, and about 0.01w/v% to 0.15w/v% polysorbate 20.
According to one embodiment, the pharmaceutical composition has a pH of about 5 to about 6 (e.g., about 5.5).
According to one embodiment, the pharmaceutical composition does not contain any cryoprotectant, such as a sugar or sugar alcohol.
According to one embodiment, the radioactive metal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 350 μci per about 2mg of total antibody when administered.
According to one embodiment, the pharmaceutical composition comprises a total amount of about 0.1mg/mL to 1.0mg/mL of the conjugate intermediate and the radioactive conjugate; for example, about 0.5mg/mL.
Another embodiment of the present invention provides a method of preparing a pharmaceutical composition comprising combining a first intermediate composition and a second intermediate composition to form the pharmaceutical composition, wherein: the first intermediate composition comprises a radioactive conjugate and the second intermediate composition comprises a conjugate intermediate and does not comprise any radioactive conjugate.
Drawings
The following drawings form a part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the description of specific embodiments presented herein.
FIGS. 1A and 1B are high performance liquid chromatograms (FIG. 1A) and of pharmaceutical product compositions 111 High performance liquid chromatography (FIG. 1B) of In-DOTA-h11B 6.
FIGS. 2A and 2B are 225 High performance liquid chromatography of Ac-DOTA-h11B6 pharmaceutical product comprising sucrose and lacking ascorbate at t=0 (fig. 2A) and t=96 hours (fig. 2A).
Fig. 3A to 3D are high performance liquid chromatograms of a pharmaceutical product. The composition contained about 50 μCi (FIGS. 3A and 3B) and 200 μCi per 4mL of drug product 225 Ac-DOTA-h11B6 (FIG. 3C and FIG. 3D). The compositions of fig. 3A and 3C further comprise sodium ascorbate, and the compositions of fig. 3B and 3D further comprise sucrose.
Fig. 4A and 4B show examples of radioactive conjugates of the present invention (bond between Ac-225 and chelator not shown).
Fig. 5 shows an example of a conjugate intermediate of the invention. Figure 5A provides an illustration of a conjugate intermediate comprising DOTA, wherein the lysine moiety is not shown. FIG. 5B provides a schematic representation of a conjugate intermediate comprising DOTA, wherein the lysine moiety is represented. Figure 5C provides an illustration of the conjugation process of a conjugate intermediate comprising DOTA.
FIG. 6 shows an example of a conjugate intermediate of the invention (TOPA-h 11B 6). Figure 6A provides an illustration of a conjugate intermediate comprising a TOPA chelator (TOPA-h 11B 6), where the lysine moiety is not shown. FIG. 6B provides a schematic representation of the conjugate intermediate shown in FIG. 6A, wherein the lysine moiety is represented. Fig. 6C provides an illustration of the conjugation process of the conjugate intermediate.
FIG. 7 shows the amino acid sequences of the heavy and light chains of the h11B6 antibody.
Detailed Description
The compounds, compositions and methods described herein are useful for treating cancer in a patient in need of such treatment. Embodiments of the compounds, compositions, and methods are effective in treating prostate cancer, including advanced prostate cancer, particularly castration-resistant prostate cancer (CRPC), and thus increasing survival in patients. These compounds are particularly useful in patients where existing treatments for advanced prostate are considered unsuccessful.
Human kallikrein 2 (hK 2) is a trypsin-like antigen produced by columnar prostate epithelial cells and driven by Androgen Receptor (AR) signaling in the same manner as closely related prostate specific antigen (PSA; human glandular kallikrein 3), which has 80% homology genetically to the PSA gene. However, unlike PSA, circulating levels of hK2 were found to be at abnormally low levels, where they could be bound by a variety of protease inhibitor complexes. Although hK2 is thought to be mainly secreted, there is evidence that it is able to induce internalization via antigen-antibody complexes, and thus it is thought to also be present on the cell surface. Because hK2 expression is highly specific for prostate adenocarcinoma and increases throughout disease progression, therapies targeting hK2 are attractive.
Exemplary hK2 sequences are described as transcripts: KLK2-201 (ENST 00000325321), provided herein as SEQ ID NO:7, the product of gene ENSG00000167751, as given in the general database.
Certain terms
An "antigen binding fragment" or "antigen binding domain" refers to that portion of an isolated protein that binds an antigen. The antigen binding domain may be a synthetic, enzymatically obtainable or genetically engineered polypeptide and includes antigen-binding portions of immunoglobulins such as VH, VL, VH and VL, fab, fab ', F (ab') 2 Fd and Fv fragments; a domain antibody (dAb) consisting of one VH domain or one VL domain; a shark variable IgNAR domain; humping the VH domain; a VHH domain; from CDRs of a mimetic antibody, e.g., FR3-CDR3-FR4 portions, HCDR1, HCDR2 and/or HCDR3, LCDR1, LCDR2 and/or LCDR 3; an alternative scaffold that binds antigen; and multispecific proteins comprising antigen-binding fragments. Antigen binding fragments, such as VH and VL, can be linked together via synthetic linkers to form various types of single antibody designs, wherein in those cases where the VH and VL domains are expressed from separate single chains, the VH/VL domains can be paired intramolecularly or intermolecularly to form monovalent antigen binding sites, such as single chain Fv (scFv) or diabodies.
"antibody" broadly refers to and includes immunoglobulin molecules, particularly including monoclonal antibodies (including murine monoclonal antibodies, human monoclonal antibodies, humanized monoclonal antibodies, and chimeric monoclonal antibodies), antigen binding fragments, multispecific antibodies (such as bispecific antibodies, trispecific antibodies, tetraspecific antibodies), dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies, and any other modified configuration of immunoglobulin molecules comprising an antigen binding site having the desired specificity. "full length antibodies" comprise two Heavy Chains (HC) and two Light Chains (LC) interconnected by disulfide bonds and multimers thereof (e.g., igM). Each heavy chain consists of a heavy chain variable region (VH) and a heavy chain constant region (consisting of domains CH1, hinge, CH2 and CH 3). Each light chain is composed of a light chain variable region (VL) and a light chain constant region (CL). The VH and VL regions may be further subdivided into regions of hypervariability termed Complementarity Determining Regions (CDRs) interspersed with Framework Regions (FR). Each VH and VL is made up of three CDRs and four FR segments, and arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. Immunoglobulins can be assigned to five major classes, igA, igD, igE, igG and IgM, based on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified into isotypes IgA1, igA2, igG1, igG2, igG3 and IgG4. Based on the amino acid sequence of its constant domain, the antibody light chain of any spinal species can be assigned to one of two completely different types, namely kappa and lambda.
The term "variant" when used in relation to an antigen or antibody may refer to a peptide or polypeptide comprising one or more (such as, for example, from about 1 to about 25, from about 1 to about 20, from about 1 to about 15, from about 1 to about 10, or from about 1 to about 5) amino acid sequence substitutions, deletions, and/or additions as compared to the native or unmodified sequence. For example, hK2 variants may result from one or more (such as, for example, from about 1 to about 25, from about 1 to about 20, from about 1 to about 15, from about 1 to about 10, or from about 1 to about 5) changes to the amino acid sequence of native hK 2. Also by way of example, variants of an anti-hK 2 antibody (such as h11B 6) may be derived from changes to one or more (such as, for example, from about 1 to about 25, from about 1 to about 20, from about 1 to about 15, from about 1 to about 10, or from about 1 to about 5) of the amino acid sequence of a natural or previously unmodified anti-hK 2 antibody. Variants may be naturally occurring, such as alleles or splice variants, or may be artificially constructed. Polypeptide variants can be prepared from the corresponding nucleic acid molecules encoding the variants. In specific embodiments, the hK2 variant or anti-hK 2 antibody variant retains at least the functional activity of hK2 or anti-hK 2 antibody, respectively. In specific embodiments, the anti-hK 2 antibody variant binds to hK2 and/or antagonizes hK2 activity. In certain embodiments, the variant is encoded by a Single Nucleotide Polymorphism (SNP) variant of a nucleic acid molecule encoding an hK2 or anti-hK 2 antibody VH or VL region or sub-region, such as one or more CDRs.
When used with respect to antibodies, a non-limiting example of a variant is an "Fc variant," which is an antibody having a variant Fc region. A "variant Fc region" includes an amino acid sequence that differs from the amino acid sequence of a native sequence Fc region by at least one amino acid modification (e.g., substitution, addition, or deletion). In certain embodiments, the variant Fc region has at least one amino acid substitution compared to the native sequence Fc region or the Fc region of the parent polypeptide, e.g., about one to about ten amino acid substitutions, or about one to about five amino acid substitutions, in the native sequence Fc region or the Fc region of the parent polypeptide. The variant Fc-regions herein may have at least about 80% homology with the native sequence Fc-region and/or the Fc-region of the parent polypeptide, or at least about 90% homology therewith, e.g., at least about 95% homology therewith.
The term "identity" refers to the relationship between sequences of two or more polypeptide molecules or two or more nucleic acid molecules as determined by aligning and comparing sequences. "percent (%) sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical to amino acid residues in the reference polypeptide sequence, without regard to any conservative substitutions as part of the sequence identity after aligning the sequences and introducing gaps (if desired) to achieve the maximum percent sequence identity. Alignment for the purpose of determining percent amino acid sequence identity can be accomplished in a variety of ways within the skill of the art, for example using publicly available computer software such as BLAST, BLAST-2, ALIGN, or megasign (DNAStar, inc.) software. One skilled in the art can determine the appropriate parameters for aligning sequences, including any algorithms needed to achieve maximum alignment over the full length of the compared sequences.
"specific binding" or "binding" refers to the binding of a protein molecule to an antigen or epitope within the antigen with greater affinity than to other antigens. Typically, protein molecules bind to an antigen or epitope within an antigen, equilibrium dissociation constants (K D ) Is about 1X 10 -7 M or less, e.g. about 5X 10 -8 M or less, about 1X 10 -8 M or less, about 1X 10 -9 M or less, about 1X 10 -10 M or less, about 1X 10 -11 M or less or about 1X 10 -12 M or less, usually K D K in comparison to its binding to non-specific antigens (e.g. BSA, casein) D At least one hundred times lower. As used herein, an antibody or antigen binding domain that "has binding specificity for hK 2" refers to an antibody or antigen binding domain, respectively, that specifically binds to hK 2.
As used herein, in certain embodiments, the term "subject" refers to a mammal, such as a non-primate (e.g., cow, pig, horse, cat, dog, rat, etc.) or primate (e.g., monkey and human). In particular embodiments, the subject is a human. In one embodiment, the subject is a mammal, such as a human, diagnosed with a condition or disorder. In another embodiment, the subject is a mammal, such as a human, at risk of developing a condition or disorder. As used herein, the term "patient" refers to a human.
"administration" refers to the act of physically delivering a substance present in vitro into a patient by injection or otherwise, such as by mucosal, intradermal, intravenous, intramuscular, subcutaneous delivery, and/or any other physical delivery method described herein or known in the art.
As used herein, the term "treatment" refers to a reduction or improvement in the progression, severity, and/or duration of a disease or disorder caused by administration of one or more therapies. Treatment may be determined by assessing whether there has been a reduction, alleviation and/or relief of one or more symptoms associated with the underlying disorder such that an improvement in the patient is observed despite the patient possibly still being afflicted with the underlying disorder. The term "treatment" includes management and amelioration of the disease. The term "management" (manage, managing and management) refers to the beneficial effect a subject obtains from a therapy that does not necessarily result in cure of a disease.
As used herein, the term "effective amount" or "therapeutically effective amount" refers to an amount of a radio conjugate or pharmaceutical composition provided herein sufficient to produce a desired therapeutic effect for a given condition and administration regimen.
The terms "agent," "drug" (pharmaceutical, drug, medication), "active agent," "active pharmaceutical ingredient" (API), and "active" are used interchangeably herein to describe a pharmaceutically active compound in a pharmaceutical composition. An example of an API suitable for use in accordance with the present invention is a radioactive conjugate having binding specificity for hK 2. The pharmaceutical composition may comprise one or more APIs and one or more additional ingredients referred to herein as "excipients". Preferably, the excipient is substantially or entirely pharmaceutically inert.
The term "dose" refers to the total amount of a particular pharmaceutical composition administered to a patient at a particular time. Preferably, one dose is delivered in the form of a single administration unit dose of the pharmaceutical composition (e.g., via intravenous administration). Alternatively, a unit dose that has been subdivided into a plurality of sub-doses (where sub-doses refer to a portion of the unit dose) may be administered multiple times.
As used herein, the term "pharmaceutically acceptable" means a substance that is non-toxic and preferably permitted by regulatory authorities (e.g., the european or federal or state government) or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
Radioactive decay refers to the process by which an unstable nucleus loses energy by radiation to produce at least one daughter nuclide. Half-life refers to the time required for half of the nuclei of a radioactive sample to decay to its daughter nuclide. The non-international unit system of measurement of radioactivity of a substance is curie (Ci). A Curie equal to 37,000,000,000 (3.7X10) of atoms decaying per second 10 ) Is used as a radioactive material. Another unit of measure of radioactivity of a substance is the International unit system of Beckler (Bq). Beckle is equal to the amount of radioactive material in which one nucleus per second decays. Specific activity refers to the amount of radioactivity per unit mole or mass in a sample; for example, it is sometimes expressed as Ci/mmol or Ci/mg. The radioactivity concentration, also referred to as specific concentration (e.g., expressed as mCi/mL or μci/mL), refers to the total amount of radioactivity per unit volume.
With respect to immunoconjugates and radio-conjugates, the term "conjugated" means "conjugated". Molecules (such as antibodies and chelators) may be attached to each other, for example, by covalent bonding.
"cancer" refers to a wide variety of diseases characterized by uncontrolled growth of abnormal cells in the body. Uncontrolled cell division and growth results in the formation of malignant tumors that invade adjacent tissues, and may also metastasize to distal parts of the body through the lymphatic system or blood flow. "cancer" or "cancer tissue" may include tumors.
The transitional term "comprising" is intended to imply its well-known meaning in the patent literature. "comprising" is synonymous with "including" or "containing" and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.
The term "between" as used in phrases such as "between a and B" or "between a-B" refers to a range that includes both a and B. The term "slave" as used in phrases such as "from a to B" or "from a-B" refers to a range that includes both a and B.
When values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms another embodiment. Generally, the use of the term "about" refers to approximations that may vary depending upon the desired properties sought to be obtained by the disclosed subject matter, and will be explained based on their function in the specific context in which the approximation is used. In some cases, the number of significant digits for a particular value can be one non-limiting method of determining the scope of the word "about". In other cases, gradients used in a series of values may be used to determine the expected range of values for each that can be used for the term "about. Where present, all ranges are inclusive and combinable. That is, a reference to a value specified in a range includes every value within that range. In certain embodiments, the term "about" means a variance of ±10% of the correlation value, and additional embodiments include those wherein the variance may be ±5%, ±15%, ±20%, ±25% or ±50%.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. That is, each individual embodiment is considered combinable with any other embodiment, and such combination is considered another embodiment, unless expressly incompatible or explicitly excluded. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any subcombination. Finally, although an embodiment may be described as part of a series of steps or as part of a more general structure, each of the steps may itself be considered a separate embodiment, which may be combined with other embodiments.
When a list is provided, it is to be understood that each individual element in the list and each combination of the list is a separate embodiment unless indicated otherwise. For example, a list of embodiments presented as "A, B or C" will be understood to include embodiments "a", "B", "C", "a or B", "a or C", "B or C" or "A, B or C".
Abbreviations used in this disclosure include
The present invention may be understood more readily by reference to the following description of the entire figures and examples, which form a part of this disclosure. It is to be understood that this invention is not limited to the specific compounds, methods, conditions or parameters described or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of any claimed invention. Similarly, any description of possible mechanisms or modes of action or reasons for improvement is intended for illustrative purposes only, and the invention herein is not limited by the correctness or mistakes of any such suggested mechanisms or modes of action or reasons for improvement, unless specifically stated otherwise.
The radioactive conjugate of the present invention
Embodiments of the present invention relate to compositions and methods for targeting hK2 with a radioactive conjugate to achieve effective cancer cell death (e.g., tumor cell death) in prostate cancer patients. As used herein, "immunoconjugate" refers to an antibody or antigen binding domain conjugated (e.g., conjugated via a covalent bond) to a second molecule such as a toxin, drug, radiometal ion, chelator, radiometal complex, or the like. "radio conjugate" (also referred to herein as a "radio immunoconjugate") particularly refers to an antibody or antigen binding domain conjugated (e.g., conjugated via a covalent bond) to at least one radio-metal complex. In other words, a radioconjugate refers to at least one radiometal complex that is conjugated (e.g., bound via a covalent bond) to an antibody or antigen binding domain. The radioconjugate may comprise at least one radiometal complex comprising a linker, wherein the radiometal complex is conjugated to the antibody or antigen binding domain via the linker.
As used herein, "antibody-chelator complex" or "conjugate intermediate" or "drug substance intermediate" refers to a precursor of a radioactive conjugate that comprises an antibody or antigen binding domain conjugated (e.g., conjugated via a covalent bond) to a chelator that does not comprise a radioactive metal. The conjugate intermediate may comprise a linker, wherein the chelator is conjugated to the antibody or antigen binding domain via the linker. After the radiometal is chelated to the chelator of the conjugate intermediate, it becomes a radioconjugate. For example, "DOTA-mAb" refers to a conjugate intermediate comprising DOTA conjugated to an antibody. An example of a conjugate intermediate is DOTA-h11B6. As used herein, "DOTA-h11B6" is a conjugate intermediate comprising DOTA conjugated to h11B6, optionally via a linker. Non-limiting examples of DOTA-mAbs are shown in FIGS. 5A-5C. Another example of a conjugate intermediate is TOPA-h11B6. As used herein, "TOPA-h11B6" is a conjugate intermediate comprising TOPA conjugated to h11B6, optionally via a linker. Non-limiting examples of TOPA-mAbs are shown in FIGS. 6A-6C.
The chelator may be conjugated to the antibody according to methods known in the art; for example, the chelator may be conjugated to the antibody via a linker. Thus, the radioactive conjugates and conjugate intermediates of the invention may comprise a chelator conjugated to the antibody through a linker. As used herein, the term "linker" generally refers to a chemical moiety that binds a chelator to an antibody or antigen binding domain. Any suitable linker known to those skilled in the art may be used in the present invention in light of the present disclosure. The linker may comprise, for example, a substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl moiety, substituted or unsubstituted aryl or heteroaryl, a polyethylene glycol (PEG) linker, a peptide linker, a sugar-based linker, or a cleavable linker, such as a disulfide bond or a protease cleavage site, such as valine-citrulline-p-aminobenzyl (PAB).
According to certain embodiments, the chelator or chelator-linker comprises a nucleophilic moiety or electrophilic moiety as described herein. The reaction of the nucleophilic or electrophilic group of the chelator or chelator-linker with the antibody or antigen-binding domain comprising the corresponding reaction partner allows covalent bonding of the antibody or antigen-binding domain to the chelator-linker. As used herein, with respect to compounds of formulae (I), (II), (III), (IV), (V) and (VI), the linker (L 1 ) Can be associated with an electrophilic or nucleophilic moiety (R 11 ) Joining to form-L 1 -R 11 . Examples of nucleophilic groups include, but are not limited to, azides, amines, and thiols. Examples of electrophilic groups include, but are not limited to, amine-reactive groups, thiol-reactive groups, alkynyl groups, and cycloalkynyl groups. The amine reactive groups preferably react with primary amines, including primary amines present in the N-terminus of each polypeptide chain and in the side chains of lysine residues. Examples of amine reactive groups include, but are not limited to, N-hydroxysuccinimide (NHS), substituted NHS (such as sulfo-NHS), isothiocyanate (-NCS), isocyanate (-NCO), ester, carboxylic acid, acid halide, amide, alkylamide, and tetrafluorophenyl and perfluorophenyl esters. The thiol reactive group reacts with a thiol or thiol group (preferably a thiol present in the side chain of a cysteine residue of the polypeptide). Examples of thiol-reactive groups include, but are not limited to, michael acceptors (e.g., maleimides), haloacetyl groups, acid halides, activated disulfides, and phenyloxadiazole sulfones.
According to certain embodiments, the conjugation reaction results in the addition of one or more chelator molecules (e.g., DOTA molecules) to the epsilon amino groups of the lysine side chains of an antibody (e.g., h11B6 mAb). For example, 1, 2, 3, 4, or 5 DOTA molecules may be conjugated to an antibody. According to certain embodiments, p-SCN-Bn-DOTA may be reacted with an antibody to form a conjugate intermediate comprising DOTA, as shown in fig. 5. Figure 5A provides an illustration of a conjugate intermediate comprising DOTA, wherein the lysine moiety is not shown. FIG. 5B provides a schematic representation of a conjugate intermediate comprising DOTA, wherein the lysine moiety is represented. Fig. 6B and 6C provide illustrations of conjugate intermediates comprising alternative chelators according to the invention.
The chelator to antibody ratio (CAR), which represents the number of chelator-linker molecules per antibody molecule, can be measured by complete mass analysis using RP-HPLC with online mass analysis. According to certain embodiments, the average CAR of a conjugate intermediate of the invention (e.g., DOTA-mAb such as DOTA-h11B 6) is from about 1 to about 8, from about 1 to about 7, or from about 1 to about 6, or from about 1 to about 5, or from about 1 to about 4, or from about 1 to about 3, or from about 2 to about 4, or from about 2 to about 3.
According to a particular embodiment, the radioconjugates described herein comprise a radiometal complex conjugated to an antibody or antigen binding domain having binding specificity for kallikrein related peptidase 2 (hK 2). According to a particular embodiment, the radioactive conjugate is a radiolabeled antibody comprising an antibody conjugated (conjugated) to a radioactive metal complex. According to a particular embodiment, the radioactive conjugate comprises an antibody, such as h11B6, conjugated to a radioactive metal complex comprising a chelator and a radioactive metal. In some embodiments, the antibody is covalently bound to a chelator. The radiometal complex optionally comprises a linker, as described herein.
As used herein, "radiometal complex" refers to a complex comprising a radiometal ion associated with a chelator that is a macrocyclic compound. Typically, the radioactive metal ion is bound or coordinated to the macrocyclic compound via a coordinate bond. The heteroatoms of the macrocycle may be involved in coordinate bonding of the radioactive metal ion to the macrocyclic compound. The macrocyclic compound may be substituted with one or more substituent groups, and the one or more substituent groups may also participate in coordination bonding of the radiometal ion to the macrocyclic compound, in addition to or alternatively to the heteroatom of the macrocyclic compound. Other examples of possible linkages between chelators and radioisotopes include guest-host binding such as ionic bonding, hydrogen bonding, valance forces, or hydrophobic interactions. The radiometal complex may optionally comprise a linker which is a chemical moiety that binds the chelator to the antibody or antigen binding domain.
As used herein, the terms "radiometal," "radioisotope," "radiometal ion," and "radiometal ion (radioactive metal ion)" are used interchangeably and refer to one or more isotopes of an element that emits particles and/or photons. Non-limiting examples of radioisotopes useful in therapeutic applications according to the present invention include, for example, beta emitters or alpha emitters, such as, for example 225 Ac、 177 Lu、 32 P、 47 Sc、 67 Cu、 77 As、 89 Sr、 90 Y、 99 Tc、 105 Rh、 109 Pd、 111 Ag、 131 I、 134 Ce、 149 Tb、 152 Tb、 155 Tb、 153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 255 Fm sum 227 Th. Other non-limiting examples of radioisotopes useful as imaging agents according to the present invention include gamma-emitting radioisotopes such as, for example 177 Lu、 62 Cu、 64 Cu、 67 Ga、 68 Ga、 86 Y、 89 Zr and 111 in. In certain embodiments, the radioactive metal ion is a "therapeutic emitter," meaning a radioactive metal ion that can be used in therapeutic applications. Examples of therapeutic emitters include, but are not limited to, beta emitters or alpha emitters, such as 132 La、 135 La、 134 Ce、 144 Nd、 149 Tb、 152 Tb、 155 Tb、 153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 177 Lu、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 225 Ac、 255 Fm, and a method for producing the same 227 Th、 226 Th、 230 U, U. Preferably, the radioactive metal ions used in the present invention are alpha-emitting radioactive metal ions, such as actinium-225 # 225 Ac)。
Note that certain radioactive metals may be used as therapeutic agents (e.g., 225 ac) and/or an imaging agent (e.g., 111 in). Suitable radiometals for use as therapeutic agents are radiometals capable of reducing or inhibiting the growth of, or in particular killing, cancer cells, such as prostate cancer cells. In certain embodiments, the radioactive conjugates of the present invention can deliver a cytotoxic payload that has the ability to emit alpha and/or beta particles near a tumor by binding to a cancer cell surface antigen and triggering cell death. In certain embodiments, the radioactive conjugate of the invention is internalized into a cancer cell expressing hk 2.
As used herein, the terms "225Ac", " 225 Ac "or" Ac-225 "refers to actinium-225, which is a radioactive metal that emits alpha. According to a specific embodiment of the present invention, 225 the approximately ten-day half-life of Ac (approximately 9.9 days) is long enough to be able to prepare the compounds described herein, but short enough to match the circulating pharmacokinetics of antibodies conjugated to radioactive metal complexes (such as h11B 6). 225 Ac decays in a series of steps, which reach a stable isotope 209 Bi was preceded by eventually emitting four alpha particles, providing increased compound efficacy.
Antibodies of the invention
According to a particular embodiment, the radioactive conjugate of the invention comprises an antibody, which is an h11B6 antibody. Embodiments of the h11B6 antibodies are described in U.S. patent 10,100,125, which is incorporated herein by reference. As used herein, "h11B6 antibody" or "h11B6 mAb" or "h11B6" or "hu11B6" refers to an antibody having binding specificity for human kallikrein-2 (hK 2), wherein the antibody comprises (a) a heavy chain variable region comprising the amino acid sequences of SEQ ID NO:1 and SEQ ID NO:2 and SEQ ID NO:3 and/or (B) a light chain variable region comprising the amino acid sequences of SEQ ID NO:4 and SEQ ID NO:5 and SEQ ID NO:6, wherein the heavy chain variable region and the light chain variable region comprise the framework amino acid sequences from one or more human antibodies.
According to a particular embodiment, the radioactive conjugate of the present invention comprises an h11B6 antibody comprising (a) a heavy chain variable region (VH) comprising VH CDR1 having the amino acid sequence of SEQ ID NO:1 (SDYAWN), VH CDR2 having the amino acid sequence of SEQ ID NO:2 (YISYSGSTTYNPSLKS) and VH CDR3 having the amino acid sequence of SEQ ID NO:3 (gyygsgf); and (b) a light chain variable region (VL) comprising a VL CDR1 having the amino acid sequence of SEQ ID NO. 4 (KASESVEYFGTSLMH), a VL CDR2 having the amino acid sequence of SEQ ID NO. 5 (AASNRES) and a VL CDR3 having the amino acid sequence of SEQ ID NO. 6 (QQTRKVPYT).
The six amino acid sequences described above represent Complementarity Determining Regions (CDRs) as defined according to Kabat et al, (1991) Sequences of Immunological Interest, 5 th edition, NIH, bethesda, md. (the disclosure of which is incorporated herein by reference). The Kabat numbering scheme is used throughout this specification (Kabat et al, 1991).
According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain variable region (VH) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID NO:8 and/or a light chain variable region (VL) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID NO: 9.
According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO. 8 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO. 9.
SEQ ID NO. 8 is as follows:
QVQLQESGPGLVKPSDTLSLTCAVSGNSITSDYAWNWIRQPPGKGLEWIGYISYSGSTTYNPSLKSRVTMSRDTSKNQFSLKLSSVTAVDTAVYYCATGYYYGSGFWGQGTLVTVSS
SEQ ID NO 9 is as follows:
DIVLTQSPDSLAVSLGERATINCKASESVEYFGTSLMHWYQQKPGQPPKLLIYAASNRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQTRKVPYTFGQGTKLEIK
according to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 10 and/or a light chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 11.
According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain constant region comprising the amino acid sequence of SEQ ID NO. 10 and/or a light chain constant region comprising the amino acid sequence of SEQ ID NO. 11.
SEQ ID NO 10 is as follows:
A S T K G P S V F P L A P S S K S T S G G T A A L G C L V K D Y F P E P V T V S W N S G A L T S G V H T F P A V L Q S S G L Y S L S S V V T V P S S S L G T Q T Y I C N V N H K P S N T K V D K K V E P K S C D K T H T C P P C P A P E L L G G P S V F L F P P K P K D T L M I S R T P E V T C V V V D V S H E D P E V K F N W Y V D G V E V H N A K T K P R E E Q Y N S T Y R V V S V L T V L H Q D W L N G K E Y K C K V S N K A L P A P I E K T I S K A K G Q P R E P Q V Y T L P P S R E E M T K N Q V S L T C L V K G F Y P SD I A V E W E S N G Q P E N N Y K T T P P V L D SD G S F F L Y S K L T V D K S R W Q Q G N V F S C S V M H E A L H N H Y T Q K S L S L S P G K
SEQ ID NO. 11 is as follows:
R T V A A P S V F I F P P SD E Q L K S G T A S V V C L L N N F Y P R E A K V Q W K V D N A L Q S G N S Q E S V T E Q D S K D S T Y S L S S T L T L S K A D Y E K H K V Y A C E V T H Q G L S S P V T K S F N R G E C
according to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity with the amino acid sequence of SEQ ID No. 12 and/or a light chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity with the amino acid sequence of SEQ ID No. 13.
According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO. 12 and/or a light chain having the amino acid sequence of SEQ ID NO. 13.
The amino acid sequences of the h11B6 heavy and light chains are also shown in fig. 7.
According to a particular embodiment, the antibodies of the invention (e.g. h11B 6) comprise or consist of intact (i.e. complete) antibodies, such as IgA, igD, igE, igG or IgM molecules.
According to a particular embodiment, the antibody of the invention (e.g. h11B 6) comprises or consists of an intact IgG molecule or variant thereof. The IgG molecules may be of any known subtype, such as IgG1, igG2, igG3 or IgG4.
According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody, which is an IgG1 antibody. According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody, which is an IgG1 kappa isotype. According to a particular embodiment, the radioactive conjugate of the invention comprises an h11B6 antibody, which is an IgG1 antibody or variant thereof, such as an Fc variant.
According to one embodiment, the radioactive conjugate of the invention comprises an antibody conjugated to DOTA, optionally via a linker. For example, the h11B6 antibody can be conjugated to DOTA to produce a DOTA-h11B6 conjugate intermediate, and then the DOTA-h11B6 is sequestered to 225 Ac to produce radioconjugates 225 Ac-DOTA-h11B6。
According to certain embodiments, DOTA-h11B6 is formed by chemically conjugating h11B6 with the DOTA derivative p-SCN-Bn-DOTA (CAS registry number 127985-74-4; chemical name 2-S- (4-isothiocyanatobenzyl) -1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid) according to known methodsSo that multiple DOTA molecules are added to the epsilon amino groups of the lysine side chains of the h11B6 mAb. DOTA-h11B6 can then be sequestered to 225 Ac to produce radioconjugates 225 Ac-DOTA-h11B6. When administered to a patient, the radioactive conjugate 225 Ac-DOTA-h11B6 can bind to and internalize within hK2 expressing cells.
According to one embodiment, the radioactive conjugate of the invention comprises an antibody conjugated to TOPA, optionally via a linker. For example, the h11B6 antibody can be conjugated to TOPA to generate a TOPA-h11B6 conjugate intermediate, and then the TOPA-h11B6 is sequestered to 225 Ac to produce radioconjugates 225 Ac-TOPA-h11B6。
According to certain embodiments, TOPA-h11B6 is formed as described in WO 2020/229974 or PCT/IB 2021/060350. TOPA-h11B6 can then be sequestered to 225 Ac to produce radioconjugates 225 Ac-TOPA-h11B6. When administered to a patient, the radioactive conjugate 225 Ac-TOPA-h11B6 can bind to and internalize within hK2 expressing cells.
According to one embodiment, antibodies of the invention, such as h11B6 antibodies, can be prepared as described in U.S. patent nos. 10,100,125 and 9,873,891, both of which are incorporated herein by reference. In some embodiments, the antibodies of the invention, such as h11B6, are prepared using CHO-DG44 cells.
Methods for producing antibodies are well known in the art. For example, suitable methods for producing recombinant polypeptides are known in the art, such as expression in prokaryotic or eukaryotic host cells (see, e.g., sambrook & Russell,2000, molecular Cloning, A Laboratory Manual, third edition, cold Spring Harbor, n.y., the relevant disclosure of which is incorporated herein by reference).
In one aspect, the invention provides an isolated nucleic acid molecule encoding an antibody or component polypeptide chain thereof of the invention. "nucleic acid molecules" include DNA (e.g., genomic DNA or complementary DNA) molecules and mRNA molecules, which may be single-stranded or double-stranded. In one embodiment, the nucleic acid molecule is a cDNA molecule. It will be appreciated by those skilled in the art that the nucleic acid molecules may be codon optimized to express the antibody polypeptide in a particular host cell, for example in a human cell (see, e.g., angov,2011, biotechnol. J.6 (6): 650-659).
In a particular embodiment, the nucleic acid molecule of the invention comprises (a) the nucleotide sequence of SEQ ID NO. 14 and/or (b) the nucleotide sequence of SEQ ID NO. 15.
Antibody variants of the invention
In some embodiments, amino acid sequence modifications of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of an antibody, including but not limited to specificity, thermostability, expression level, effector function, glycosylation (e.g., fucosylation), reduced immunogenicity, or solubility. Thus, it is contemplated that antibody variants may be prepared in addition to the antibodies described herein. For example, antibody variants may be prepared by introducing appropriate nucleotide changes into the encoding DNA and/or by synthesizing the desired antibody or polypeptide. Those skilled in the art will appreciate that amino acid changes may alter the post-translational processes of the antibody, such as altering the number or position of glycosylation sites or altering membrane anchoring characteristics.
In some embodiments, the antibodies provided herein are chemically modified, for example, by covalently linking any type of molecule to the antibody. Antibody derivatives may include antibodies that have been chemically modified, for example, by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, attachment to cellular ligands or other proteins, and the like. Any of a number of chemical modifications may be made by known techniques including, but not limited to, specific chemical cleavage, acetylation, formulation, metabolic synthesis of tunicamycin, and the like. In addition, antibodies may contain one or more atypical amino acids.
A variant may be a substitution, deletion, or insertion of one or more codons encoding an antibody or polypeptide that results in a change in the amino acid sequence as compared to the native sequence antibody or polypeptide. Amino acid substitutions may be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as replacing leucine with serine, e.g., a conservative amino acid substitution. Standard techniques known to those skilled in the art can be used to introduce mutations in the nucleotide sequences encoding the molecules provided herein, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis resulting in amino acid substitutions. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. In certain embodiments, substitutions, deletions, or insertions include less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original molecule. In a specific embodiment, the substitution is a conservative amino acid substitution at one or more predicted nonessential amino acid residues. The allowed variants can be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full length or mature native sequence.
Amino acid sequence insertions include amino and/or carboxy terminal fusions ranging in length from one residue to polypeptides containing one hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include antibodies with an N-terminal methionyl residue. Other insertional variants of antibody molecules include fusions of the N-terminus or C-terminus of an antibody with an enzyme (e.g., for antibody-directed enzyme prodrug therapy) or polypeptide, which increases the serum half-life of the antibody.
A "conservative amino acid substitution" is a substitution in which an amino acid residue is replaced with an amino acid residue having a side chain with a similar charge. The art has defined families of amino acid residues with similarly charged side chains. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations may be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resulting mutants may be screened for biological activity to identify mutants that retain activity. After mutagenesis, the encoded protein may be expressed and the activity of the protein may be determined.
Basic modifications in the biological properties of antibodies are achieved by selection of substitutions that maintain the structure of the (a) polypeptide backbone in the substitution region, e.g., as a folded or helical conformation; (b) the charge or hydrophobicity of the molecule at the target site; or (c) the effect of the bulk of the side chain is significantly different. Alternatively, conservative substitutions (e.g., within amino acid groups having similar properties and/or side chains) may be made in order to maintain or not significantly alter the properties. Amino acids may be grouped according to their similarity in side chain characteristics (see, e.g., lehninger,Biochemistry73-75 (2 nd edition 1975)): (1) nonpolar: ala (A), val (V), leu (L), ile (I), pro (P), phe (F), trp (W), met (M); (2) uncharged polarity: gly (G), ser (S), thr (T), cys (C), tyr (Y), asn (N), gln (Q); (3) acidity: asp (D), glu (E); and (4) alkaline: lys (K), arg (R), his (H).
Alternatively, naturally occurring residues can be divided into several groups based on common side chain characteristics: (1) hydrophobicity: norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic: cys, ser, thr, asn, gln; (3) acidity: asp, glu; (4) alkaline: his, lys, arg; (5) residues that affect chain orientation: gly, pro; and (6) aromatic: trp, tyr, phe.
Non-conservative substitutions require the exchange of members of one of these classes for another class. Such substituted residues may also be introduced into conserved substitution sites or into the remaining (non-conserved) sites.
Thus, in one embodiment, an antibody that binds an hK2 epitope comprises an amino acid sequence that is at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identical to the amino acid sequence of an antibody described herein, e.g., the h11B6 antibody described herein.
Chelating agents of the invention
According to a particular embodiment, the chelator of the present invention refers to a chelator with which a metal (preferably a radiometal) can complex to form a radiometal complex. Preferably, the chelating agent is a macrocyclic compound. In certain embodiments, the chelator comprises a macrocyclic (macro/macro ring) ring containing one or more heteroatoms (e.g., oxygen and/or nitrogen) as ring atoms.
According to a particular embodiment, the chelating agent comprises a macrocyclic chelating moiety. Examples of macrocyclic chelating moieties include, but are not limited to, 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA), S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA), 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA), 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA), 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A), or derivatives thereof. In some aspects, the chelator is 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA). In other aspects, the chelator is S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA). In other aspects, the chelator is 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA). In other aspects, the chelator is 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA). In a further aspect, the chelator is 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A). In other aspects, the chelator is DOTA, DFO, DTPA, NOTA or TETA.
In an alternative embodiment, the chelating agent comprises a macrocycle which is a derivative of 4, 13-diaza-18-crown-6. 4, 13-diaza-18-crown-6 may be in a variety of formsPreparation of formula (see, e.g., gatto et al, org.Synth.1990,68,227;DOI:10.15227/orgsyn.068.0227). According to other embodiments of the invention, the chelating agent is H 2 bp18c6 or H 2 bp18c6 derivatives such as those described in WO 2020/229974. H 2 bp18c6 means N, N' -bis [ (6-carboxy-2-pyridinyl) methyl)]-4, 13-diaza-18-crown-6, as described herein. H 2 bp18c6 and H 2 bp18c6 derivatives are also described, for example, in Thiele et al, "An weight-Membered Macrocyclic Ligand for Actinium-225Targeted Alpha Therapy"Angew.Chem.Int.Ed "(2017) 56:14712-14717, and Roca-Sabio et al," Macrocyclic Receptor Exhibiting Unprecedented Selectivity for Light Lanthanides "j.am.chem.soc. (2009) 131,3331-3341, which are incorporated herein by reference. Additional chelators suitable for use in accordance with the present invention are described in WO2018/183906 and WO2020/106886, which are incorporated herein by reference.
As used herein, the term "TOPA" refers to a polypeptide known in the art as H 2 The macrocycle of bp18c6, and may alternatively be referred to as N, N' -bis [ (6-carboxy-2-pyridinyl) methyl) ]-4, 13-diaza-18-crown-6 or is known as 6,6' - ((1,4,10,13-tetraoxa-7, 16-diazadioctadec-7, 16-diyl) bis (methylene)) pyridinedicarboxylic acid. See for example Roca-Sabio et al,
chelating agents of the formulae (I), (II) and (III)
Additional chelating agents suitable for use in accordance with the present invention are described in WO2020/229974, which is incorporated herein by reference. According to a particular embodiment, the chelating agent has the structure of formula (I), for example as described in WO 2020/229974:
wherein:
ring a and ring B are each independently a 6-10 membered aryl or a 5-10 membered heteroaryl, wherein ring a and ring B are each optionally substituted with one or more substituents independently selected from the group consisting of: halo, alkyl, alkeneRadicals, cycloalkyl radicals, cycloalkenyl radicals, aryl radicals, heterocyclic radicals, heteroaryl radicals, -OR 13 、-SR 13 、-(CH 2 ) p COOR 13 、-OC(O)R 13 、-N(R 13 ) 2 、-CON(R 13 ) 2 、-NO 2 、-CN-OC(O)N(R 13 ) 2 And X;
Z 1 and Z 2 Each independently is- (C (R) 12 ) 2 ) m -or- (CH) 2 ) n -C(R 12 )(X)-(CH 2 ) n -;
Each X is independently-L 1 -R 11
Each n is independently 0,1, 2, 3, 4, or 5;
each m is independently 1, 2, 3, 4, or 5;
each p is independently 0 or 1;
L 1 absent or a linker;
R 11 is a nucleophilic or electrophilic moiety, or R 11 Comprising an antibody or antigen binding domain;
each R 12 Independently hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl or heteroaryl;
Each R 13 Independently hydrogen or alkyl;
R 14 、R 15 、R 16 and R is 17 Each independently is hydrogen, alkyl or X,
or alternatively R 14 And R is 15 And/or R 16 And R is 17 Taken together with the carbon atom to which they are attached to form a 5-or 6-membered cycloalkyl ring optionally substituted with X;
provided that the chelating agent comprises at least one X, and when X is present on ring A or ring B, L 1 Is a linking group, or R 12 And R is 14 -R 17 At least one of which is not hydrogen.
According to an embodiment of the invention, the chelating agent comprises at least one X group, wherein X is-L 1 -R 11 Wherein L is 1 Is absent or a linker, and R 11 Is an electrophilic or nucleophilic moiety, or R 11 Comprising an antibody or antigen binding domain. When R is 11 When nucleophilic or electrophilic moieties, such moieties may be used to attach the chelator directly or indirectly via a linker to an antibody or antigen binding domain. According to a preferred embodiment, R 11 Comprising antibodies with binding specificity for hK2, such as h11B6.
In certain embodiments, the chelator comprises a single X group, and preferably L of the X group 1 Is a linking group.
The chelators of the invention may be at any of the carbon atoms of the macrocycle, Z 1 Or Z is 2 Substituted in position or on ring A or ring B by X, provided that when ring A or ring B contains an X group, L 1 Is a linking group, or R 12 And R is 14 -R 17 At least one of which is other than hydrogen (i.e. Z 1 、Z 2 At least one of the carbon atoms of the (c) and/or the carbon of the macrocycle is substituted, for example, with an alkyl group such as methyl or ethyl). Preferably, substitution at such positions does not affect the chelator to the radioactive metal ion, in particular 225 The chelation efficiency of Ac, and in some embodiments, substitution may increase the chelation efficiency.
In some embodiments, L 1 Is not present. When L 1 R in the absence of 11 Directly bind (e.g., via covalent bonding) to the chelator.
In some embodiments, L 1 Is a linking group. As used herein, with respect to compounds of formulae (I), (II), (III), (IV), (V) and (VI), L 1 Refers to the conjugation of a chelator to a nucleophilic moiety, electrophilic moiety, antibody or chemical moiety of an antigen binding domain. Any suitable linker known to those skilled in the art may be used in the present invention in light of the present disclosure. The linker may comprise, for example, a substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl moiety, substituted or unsubstituted aryl or heteroaryl, a polyethylene glycol (PEG) linker, a peptide linker, a sugar-based linker, or a cleavable linker, such as a disulfide bond or a protease cleavage site, such as valine-citrulline-p-aminobenzyl (PAB). Applicable to Exemplary linker structures include, but are not limited to:
wherein n is an integer from 0 to 10, preferably an integer from 1 to 4; and m is an integer from 0 to 12, preferably an integer from 0 to 6.
In some embodiments, R 11 Is a nucleophilic moiety or an electrophilic moiety. "nucleophilic moiety" or "nucleophilic group" refers to a functional group that provides an electron pair in a chemical reaction to form a covalent bond. "electrophilic moiety" or "electrophilic group" refers to a functional group that accepts an electron pair in a chemical reaction to form a covalent bond. The nucleophilic group reacts with the electrophilic group and vice versa to form a new covalent bond in the chemical reaction. The reaction of the nucleophilic or electrophilic group of the chelator of the invention with an antibody or antigen-binding domain or other chemical moiety (e.g., a linker) comprising a corresponding reaction partner allows covalent bonding of the antibody or antigen-binding domain or chemical moiety to the chelator of the invention.
Illustrative examples of nucleophilic groups include, but are not limited to, azides, amines, and thiols. Illustrative examples of electrophilic groups include, but are not limited to, amine-reactive groups, thiol-reactive groups, alkynyl groups, and cycloalkynyl groups. The amine reactive groups preferably react with primary amines, including primary amines present in the N-terminus of each polypeptide chain and in the side chains of lysine residues. Examples of amine-reactive groups suitable for use in the present invention include, but are not limited to, N-hydroxysuccinimide (NHS), substituted NHS (such as sulfo-NHS), isothiocyanate (-NCS), isocyanate (-NCO), ester, carboxylic acid, acid halide, amide, alkylamide, and tetrafluorophenyl and perfluorophenyl esters. The thiol reactive group reacts with a thiol or thiol group (preferably a thiol present in the side chain of a cysteine residue of the polypeptide). Examples of thiol-reactive groups suitable for use in the present invention include, but are not limited to, michael acceptors (e.g., maleimide), haloacetyl, acyl halides, activated disulfides, and phenyloxadiazole sulfones.
In particular embodiments, R 11 is-NH 2 -NCS (isothiocyanate), -NCO (isocyanate), -N 3 (azido), alkynyl, cycloalkynyl, carboxylic acid, ester, amido, alkylamide, maleimido, acyl halide, tetrazine or trans-cyclooctene, more specifically-NCS, -NCO, -N 3 Alkynyl, cycloalkynyl, -C (O) R 13 、-COOR 13 、-CON(R 13 ) 2 Maleimide groups, acid halides (e.g., -C (O) Cl, -C (O) Br), tetrazine or trans-cyclooctene, wherein each R 13 Independently hydrogen or alkyl.
In some embodiments, R 11 Is an alkynyl, cycloalkynyl, or azido group, allowing the use of click chemistry to attach chelators to antibodies or antigen binding domains or other chemical moieties (e.g., linkers). In such embodiments, the click chemistry that may be performed is azido (-N) 3 ) Huisgen cycloaddition or 1, 3-dipolar cycloaddition with alkynyl or cycloalkynyl groups to form a 1,2, 4-triazole linker or moiety. In one embodiment, the chelator comprises an alkynyl or cycloalkynyl group, and the antibody or antigen binding domain or other chemical moiety comprises an azido group. In another embodiment, the chelator comprises an azido group and the antibody or antigen binding domain or other chemical moiety comprises an alkynyl or cycloalkynyl group.
In certain embodiments, R 11 Alkynyl groups that react with azide groups, more preferably terminal alkynyl groups or cycloalkynyl groups, specifically via strain-promoted azide-alkyne cycloaddition (sparc). Examples of cycloalkynyl groups that can react with azide groups via sparc include, but are not limited to, cyclooctynyl or Bicyclononyl (BCN), cyclooctynyl Difluoride (DIFO), dibenzocyclooctynyl (DIBO), keto-DIBO, diarylazedoxynyl (BARAC), dibenzoazacyclooctynyl (DIBAC, DBCO, ADIBO), dimethoxyazacyclooctynyl (DIMAC), difluorobenzocyclooctynyl (DIFBO), monobenzocyclooctynyl (MOBO), and tetramethoxydibenzoCyclooctynyl (TMDIBO).
In a particular embodiment, R 11 Is dibenzoazacyclooctynyl (DIBAC, DBCO, ADIBO) having the following structure:
wherein R is 11 In such embodiments that are DBCO, the DBCO may be covalently linked to the chelator directly or indirectly via a linker, and preferably is attached to the chelator indirectly via a linker.
In some embodiments, R 11 Comprising an antibody or antigen binding domain. The antibody or antigen binding domain may be directly linked to the chelator via a covalent linkage or indirectly linked to the chelator via a linker. According to a preferred embodiment, R 11 Comprising antibodies with binding specificity for hK2, such as h11B6.
According to an embodiment of the invention, each of ring a and ring B is independently a 6-10 membered aryl or a 5-10 membered heteroaryl. In alternative embodiments, it is contemplated that each of ring a and ring B is an optionally substituted heterocyclyl ring, such as oxazoline. Ring a and ring B may each be optionally and independently substituted with one or more substituents independently selected from the group consisting of: halo, alkyl, alkenyl, cycloalkyl, cycloalkenyl, aryl, heterocyclyl, heteroaryl, -OR 13 、-SR 13 、-(CH 2 ) p COOR 13 、-OC(O)R 13 、-N(R 13 ) 2 、-CON(R 13 ) 2 、-NO 2 、-CN-OC(O)N(R 13 ) 2 And X. Examples of 6 to 10 membered aryl groups suitable for this purpose include, but are not limited to, phenyl and naphthyl. Examples of 5-to 10-membered heteroaryl groups suitable for this purpose include, but are not limited to, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, and imidazolyl. Examples of suitable substituents for 5-to 10-membered heteroaryl and 6-to 10-membered aryl groups include, but are not limited to, -COOH, tetrazolyl and-CH 2 COOH. In a preferred embodiment, the substituent is-COOH or tetrazolyl, which is an isostere of-COOH。
In certain embodiments, each of ring a and ring B is independently and optionally substituted with one or more carboxyl groups including, but not limited to, -COOH and-CH 2 COOH。
In certain embodiments, each of ring a and ring B is independently and optionally substituted with tetrazolyl.
In one embodiment, ring a and ring B are the same, e.g., both ring a and ring B are pyridinyl. In another embodiment, ring a and ring B are different, e.g., one of ring a and ring B is pyridinyl and the other is phenyl.
In a particular embodiment, both ring a and ring B are pyridinyl substituted with-COOH.
In a particular embodiment, both ring a and ring B are pyridinyl substituted with tetrazolyl.
In another particular embodiment, both ring a and ring B are picolinic acid groups having the structure:
according to an embodiment of the invention, Z 1 And Z 2 Each independently is- (C (R) 12 ) 2 ) m -or- (CH) 2 ) n -C(R 12 )(X)-(CH 2 ) n -; each X is independently-L 1 -R 11 The method comprises the steps of carrying out a first treatment on the surface of the Each R 12 Independently hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl or heteroaryl; each n is independently 0, 1, 2, 3, 4, or 5; and each m is independently 1, 2, 3, 4, or 5.
In some embodiments, each R 12 Independently hydrogen or alkyl, more preferably hydrogen, -CH 3 or-CH 2 CH 3
In some embodiments, each R 12 Is hydrogen.
In some embodiments, Z 1 And Z 2 Both are- (CH) 2 ) m -, wherein each m is preferably 1. In such embodiments, the carbon atom of the macrocycle, ring a or ring B is substituted with an X group.
In some embodiments, Z 1 And Z 2 One of them is- (CH) 2 ) n -C(R 12 )(X)-(CH 2 ) n -and the other is- (CH) 2 ) m -。
In some embodiments, Z 1 And Z 2 -(CH 2 ) n -C(R 12 )(X)-(CH 2 ) n One and the other of them are- (CH) 2 ) m -; each n is 0; m is 1; x is-L 1 -R 11 The method comprises the steps of carrying out a first treatment on the surface of the And L is 1 Is a linking group.
In some embodiments, Z 1 And Z 2 Both are- (CH) 2 ) m -; each m is independently 0, 1, 2, 3, 4 or 5, preferably each m is 1; and R is 14 、R 15 、R 16 And R is 17 One of them is X, and R 14 、R 15 、R 16 And R is 17 The remainder of (2) are each hydrogen.
In some embodiments, R 14 And R is 15 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl ring (i.e., cyclopentyl or cyclohexyl). Such 5-or 6-membered cycloalkyl rings may be substituted with an X group.
In some embodiments, R 16 And R is 17 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl ring (i.e., cyclopentyl or cyclohexyl). Such 5-or 6-membered cycloalkyl rings may be substituted with an X group.
In certain embodiments, the chelator has the structure of formula (II):
wherein:
A 1 is N or CR 1 Or is absent;
A 2 is N or CR 2
A 3 Is N or CR 3
A 4 Is N or CR 4
A 5 Is N or CR 5
A 6 Is N or CR 6 Or is absent;
A 7 is N or CR 7
A 8 Is N or CR 8
A 9 Is N or CR 9
A 10 Is N or CR 10
Provided that A 1 、A 2 、A 3 、A 4 And A 5 Not more than three of them are N and A 6 、A 7 、A 8 、A 9 And A 10 No more than three are N;
R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 and R is 10 Each independently selected from the group consisting of: hydrogen, halo, alkyl, alkenyl, cycloalkyl, cycloalkenyl, aryl, heterocyclyl, heteroaryl, -OR 13 、-SR 13 、-(CH 2 ) p COOR 13 、-OC(O)R 13 、-N(R 13 ) 2 、-CON(R 13 ) 2 、-NO 2 、-CN-OC(O)N(R 13 ) 2 and-a-X,
or, alternatively, any two directly adjacent R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 And R is 10 Taken together with the atoms to which they are attached to form a five-or six-membered substituted or unsubstituted carbocyclic or nitrogen-containing ring;
and Z is 1 、Z 2 、X、n、m、p、L 1 And R is 11 -R 17 As described above for formula (I),
provided that the chelating agent comprises at least one X,and when R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 And R is 10 When any one of them is X, then L 1 Is a linking group, or R 12 And R is 14 -R 17 At least one of which is not hydrogen.
In some embodiments, any two directly adjacent R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 And R is 10 Taken together with the atoms to which they are attached to form a five-or six-membered substituted or unsubstituted carbocyclic or nitrogen-containing ring. Examples of such carbocycles that may be formed include, but are not limited to, naphthyl. Examples of such nitrogen-containing rings that may be formed include, but are not limited to, quinolinyl. The carbocyclic or nitrogen-containing ring may be unsubstituted or substituted with one or more suitable substituents, e.g., -COOH, -CH 2 COOH, tetrazolyl, and the like.
In some embodiments, L 1 Is not present. When L 1 R in the absence of 11 Directly bind (e.g., via covalent bonding) to the chelator.
In some embodiments, L 1 Is a linking group. Any suitable linker known to those skilled in the art may be used in the present invention in light of the present disclosure, such as those described above.
In some embodiments, a 1 、A 2 、A 3 、A 4 And A 5 One of them is nitrogen, A 1 、A 2 、A 3 、A 4 And A 5 One of which is a carbon substituted with-COOH and the remaining of which is CH, i.e. forms a pyridinyl ring substituted with carboxylic acid.
In some embodiments, a 6 、A 7 、A 8 、A 9 And A 10 One of them is nitrogen, A 6 、A 7 、A 8 、A 9 And A 10 One of which is a carbon substituted with-COOH and the remaining of which is CH, i.e., forms a pyridinyl ring substituted with carboxylic acid.
In one embodiment,R 1 、R 2 、R 3 、R 4 And R is 5 At least one of them is-COOH. In one embodiment, R 6 、R 7 、R 8 、R 9 And R is 10 At least one of them is-COOH. In another embodiment, R 1 、R 2 、R 3 、R 4 And R is 5 At least one of them is-COOH; and R is 6, R 7 、R 8 、R 9 And R is 10 At least one of them is-COOH.
In some embodiments, a 1 And A 10 Each is nitrogen; a is that 2 Is CR (CR) 2 And R is 2 -COOH; a is that 9 Is CR (CR) 9 And R is 9 -COOH; a is that 3 -A 8 Each is CR respectively 2 、CR 3 、CR 4 、CR 5 、CR 6 、CR 7 And CR (CR) 8 The method comprises the steps of carrying out a first treatment on the surface of the And R is 3 To R 8 Each hydrogen.
In some embodiments, a 1 、A 2 、A 3 、A 4 And A 5 One of them is nitrogen, A 1 、A 2 、A 3 、A 4 And A 5 One of which is carbon substituted with tetrazolyl and the remaining of which are CH.
In some embodiments, a 6 、A 7 、A 8 、A 9 And A 10 One of them is nitrogen, A 6 、A 7 、A 8 、A 9 And A 10 One of which is carbon substituted with tetrazolyl and the remaining of which are CH.
In one embodiment, R 1 、R 2 、R 3 、R 4 And R is 5 At least one of which is tetrazolyl. In one embodiment, R 6 、R 7 、R 8 、R 9 And R is 10 At least one of which is tetrazolyl. In another embodiment, R 1 、R 2 、R 3 、R 4 And R is 5 At least one of which is tetrazolyl; and R6, R 7 、R 8 、R 9 And R is 10 At least one of which is tetrazolyl.
In some embodiments, each R 12 Is hydrogen.
In some embodiments, R 11 For alkynyl groups or cycloalkynyl groups, cyclooctyl groups or cyclooctyl derivatives, such as DBCO, are preferred.
In a particular embodiment of the chelating agent of formula (II):
A 1 and A 10 Each is nitrogen;
A 2 is CR (CR) 2 And R is 2 -COOH;
A 9 is CR (CR) 9 And R is 9 -COOH;
A 3 -A 8 each is CR respectively 2 、CR 3 、CR 4 、CR 5 、CR 6 、CR 7 And CR (CR) 8
R 3 To R 8 Each hydrogen;
Z 1 and Z 2 One of them is- (CH) 2 ) m And Z is 1 And Z 2 The other one is- (CH) 2 ) n -C(R 12 )(X)-(CH 2 ) n -;
R 12 Is hydrogen;
m is 1;
each n is 0;
x is-L 1 -R 11 Wherein L is 1 Is a linker and-R 11 Is an electrophilic group, for example, cyclooctynyl or cyclooctynyl derivative such as DBCO; and
R 14 -R 17 Each is hydrogen, or alternatively R 16 And R is 17 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl ring.
In certain embodiments, the chelator has the structure of formula (III):
wherein:
each A 11 O, S, NMe or NH, independently;
each R 18 Independently selected from the group consisting of: hydrogen, halo, alkyl, alkenyl, cycloalkyl, cycloalkenyl, aryl, heterocyclyl, heteroaryl, -OR 13 、-SR 13 、-COOR 13 、-OC(O)R 13 、-N(R 13 ) 2 、-CON(R 13 ) 2 、-NO 2 、-CN-OC(O)N(R 13 ) 2 and-a-X,
and Z is 1 、Z 2 、X、n、m、L 1 、R 11 -R 17 As described above for formula (I),
provided that the chelating agent comprises at least one X, and R 18 When X is, then L 1 Is a linking group, or R 12 And R is 14 -R 17 At least one of which is not hydrogen.
In some embodiments, each A 11 Identical, and each A 11 O, S, NMe or NH. For example, each A 11 May be S. In other embodiments, each A 11 Different, and each independently selected from O, S, NMe and NH.
In some embodiments, each R 18 Independently is- (CH) 2 ) p -COOR 13 Or tetrazolyl, wherein R 13 Is hydrogen and each p is independently 0 or 1.
In some embodiments, each R 18 COOH.
In some embodiments, each R 18 is-CH 2 COOH。
In some embodiments, each R 18 Is tetrazolyl.
In a particular embodiment of the chelating agent of formula (III):
Each R 18 COOH;
Z 1 and Z 2 One of them is- (CH) 2 ) m And Z is 1 And Z 2 The other one is- (CH) 2 ) n -C(R 12 )(X)-(CH 2 ) n -;
R 12 Is hydrogen;
m is 1; each n is 0;
x is-L 1 -R 11 Wherein L is 1 Is a linker and-R 11 Is an electrophilic group, for example cyclooctynyl or cyclooctynyl derivative such as DBCO or BCN; and
R 14 -R 17 each is hydrogen, or alternatively R 16 And R is 17 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl ring.
In a particular embodiment of the invention, the chelating agent is selected from the group consisting of:
/>
wherein:
L 1 absent or a linker;
R 11 is a nucleophilic or electrophilic moiety, or R 11 Comprising an antibody or antigen binding domain (e.g., h11B 6); and
each R 12 Independently hydrogen, -CH 3 or-CH 2 CH 3 Provided that at least one R 12 is-CH 3 or-CH 2 CH 3
In some embodiments, R 11 is-NH 2 、-NCS、-NCO、-N 3 Alkynyl, cycloalkynyl, -C (O) R 13 、-COOR 13 、-CON(R 13 ) 2 Maleimide groups, acid halides, tetrazines or trans-cyclooctenes.
In certain embodiments, R 11 Is cyclooctynyl or a cyclooctynyl derivative selected from the group consisting of: bicyclic nonynyl (BCN), a difluorinated ringOctynyl (DIFO), dibenzocyclooctynyl (DIBO), keto-DIBO, diarylazirononyl (BARAC), dibenzoazacyclooctynyl (DIBAC, DBCO, ADIBO), dimethoxyazacyclooctynyl (DIMAC), difluorobenzocyclooctynyl (DIFBO), mono-benzocyclooctynyl (MOBO) and Tetramethoxydibenzocyclooctynyl (TMDIBO).
Preferably, R 11 Is an alkynyl group or a cycloalkynyl group, more preferably a cycloalkynyl group, such as DBCO or BCN.
Exemplary chelators of the invention include, but are not limited to:
/>
such chelators may be covalently attached to an antibody or antigen binding domain to form an immunoconjugate or radioimmunoconjugate by reacting the chelator with an azide-labeled antibody or antigen binding domain to form a 1,2, 3-triazole linker via a click chemistry reaction, as described in WO 2020/229974.
The chelators of the invention may be produced by any method known in the art in accordance with the present disclosure. For example, pendant aromatic/heteroaromatic groups may be attached to the macrocyclic moiety by methods known in the art, such as those illustrated and described in WO 2020/229974.
Chelating agents of the formulae (IV), (V) and (VI)
Additional chelators suitable for use in accordance with the invention are described in PCT/IB2021/060350, which is incorporated herein by reference. According to a particular embodiment, the chelating agent has the structure of formula (IV), for example as described in PCT/IB 2021/060350:
/>
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
Alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and
R 4 is a nucleophilic moiety, an electrophilic moiety, or an antibody or antigen binding domain (e.g., h11B 6).
In some embodiments, L 1 Is not present. When L 1 R in the absence of 4 Directly to the compound (e.g., via covalent bonding).
In some embodiments, L 1 Is a linking group. As used herein, the term "linker" refers to a chemical moiety that binds a compound of the invention to a nucleophilic moiety, electrophilic moiety, or antibody or antigen binding domain. Any suitable linker known to those skilled in the art may be used in the present invention in light of the present disclosure. The linker may have, for example, a substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl moiety, substituted or unsubstituted aryl or heteroaryl, a polyethylene glycol (PEG) linker, a peptide linker, a sugar-based linker, or a cleavable linker, such as a disulfide bond or a protease cleavage site, such as valine-citrulline-p-aminobenzyl (PAB). Exemplary linker structures suitable for use in the present invention include, but are not limited to:
Wherein m is an integer from 0 to 12.
In some embodiments, R 4 Is a nucleophilic moiety or an electrophilic moiety. "nucleophilic moiety" or "nucleophilic group" refers to a functional group that provides an electron pair in a chemical reaction to form a covalent bond. "electrophilic moiety" or "electrophilic group" refers to a functional group that accepts an electron pair in a chemical reaction to form a covalent bond. The nucleophilic group reacts with the electrophilic group and vice versa to form a new covalent bond in the chemical reaction. The reaction of a nucleophilic or electrophilic group of a compound of the invention with an antibody or antigen binding domain or other chemical moiety (e.g., a linker) comprising a corresponding reaction partner allows covalent bonding of the antibody or antigen binding domain or chemical moiety to the compound of the invention.
Examples of nucleophilic groups include, but are not limited to, azides, amines, and thiols. Examples of electrophilic groups include, but are not limited to, amine-reactive groups, thiol-reactive groups, alkynyl groups, and cycloalkynyl groups. The amine reactive groups preferably react with primary amines, including primary amines present in the N-terminus of each polypeptide chain and in the side chains of lysine residues. Examples of amine-reactive groups suitable for use in the present invention include, but are not limited to, N-hydroxysuccinimide (NHS), substituted NHS (such as sulfo-NHS), isothiocyanate (-NCS), isocyanate (-NCO), ester, carboxylic acid, acid halide, amide, alkylamide, and tetrafluorophenyl and perfluorophenyl esters. The thiol reactive group reacts with a thiol or thiol group (preferably a thiol present in the side chain of a cysteine residue of the polypeptide). Examples of thiol-reactive groups suitable for use in the present invention include, but are not limited to, michael acceptors (e.g., maleimide), haloacetyl, acyl halides, activated disulfides, and phenyloxadiazole sulfones.
In certain embodiments, R 4 is-NH 2 -NCS (isothiocyanate), -NCO (isocyanate), -N 3 (azido), alkynyl, cycloalkynyl, carboxylic acid, ester, amido, alkylamide, maleimido, acyl halide, tetrazine or trans-cyclooctene, more specifically-NCS, -NCO, -N 3 Alkynyl, cycloalkynyl, -C (O) R 13 、-COOR 13 、-CON(R 13 ) 2 Maleimide groups, acid halides (e.g., -C (O) Cl, -C (O) Br), tetrazine or trans-cyclooctene, wherein each R 13 Independently hydrogen or alkyl.
In some embodiments, R 4 Is an alkynyl, cycloalkynyl, or azido group, allowing the compounds of the invention to be attached to antibodies or antigen binding domains or other chemical moieties (e.g., linkers) using click chemistry reactions. In such embodiments, the click chemistry that may be performed is azido (-N) 3 ) Huisgen cycloaddition or 1, 3-dipolar cycloaddition with alkynyl or cycloalkynyl groups to form a 1,2, 4-triazole linker or moiety. In one embodiment, the compounds of the invention comprise an alkynyl or cycloalkynyl group, and the antibody or antigen binding domain or other chemical moiety comprises an azido group. In another embodiment, the compounds of the invention comprise an azido group and the antibody or antigen binding domain or other chemical moiety comprises an alkynyl or cycloalkynyl group.
In certain embodiments, R 4 Alkynyl groups that react with azide groups, more preferably terminal alkynyl groups or cycloalkynyl groups, specifically via strain-promoted azide-alkyne cycloaddition (sparc). Examples of cycloalkynyl groups that can react with azide groups via sparc include, but are not limited to, cyclooctynyl or Bicyclononyl (BCN), cyclooctynyl Difluoride (DIFO), dibenzocyclooctynyl (DIBO), keto-DIBO, diarylazedoxynyl (BARAC), dibenzoazacyclooctynyl (DIBAC, DBCO, ADIBO), dimethoxyazacyclooctynyl (DIMAC), difluorobenzocyclooctynyl (DIFBO), monobenzocyclooctynyl (MOBO), and Tetramethoxydibenzocyclooctynyl (TMDIBO).
In certain embodiments, R 4 Is dibenzoazacyclooctynyl (DIBAC, DBCO, ADIBO) having the following structure:
wherein R is 4 In embodiments that are DBCO, DBCO may be covalently linked to the compound directly or indirectly via a linker,and is preferably indirectly attached to the compound via a linker.
In certain embodiments, R 4 Comprising an antibody or antigen binding domain. The antibody or antigen binding domain may be directly attached to the compound via a covalent linkage or indirectly attached to the compound via a linker. In a preferred embodiment, the antibody or antigen binding domain has binding specificity for hK2, such as h11B6.
In another embodiment, the chelating agent relates to a compound of formula (V):
or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and
R 4 is a nucleophilic moiety, an electrophilic moiety, or an antibody or antigen binding domain.
In another embodiment, the chelating agent is a compound of formula (VI):
or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and
R 4 is a nucleophilic moiety, an electrophilic moiety, or an antibody or antigen binding domain (e.g., h11B 6).
In another embodiment, the chelating agent is a compound as described above, wherein: r is R 1 is-L 1 -R 4 ;R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl; l (L) 1 Absent or a linker; and R is 4 Is a nucleophilic moiety, electrophilic moiety, or antibody or antigen binding domain; or a pharmaceutically acceptable salt thereof.
In another embodiment, the chelating agentIs a compound as described above, wherein R 1 Is H; r is R 2 And R is 3 Taken together with the carbon atom to which they are attached to form a quilt-L 1 -R 4 Substituted 5-or 6-membered cycloalkyl; l (L) 1 Absent or a linker; and R is 4 Is a nucleophilic moiety, electrophilic moiety, or antibody or antigen binding domain; or a pharmaceutically acceptable salt thereof:
Additional embodiments of the above chelating agents include wherein R 4 Those that are antibodies. According to a preferred embodiment, R 4 Comprising antibodies with binding specificity for hK2, such as h11B6.
In one embodiment, the chelating agent is any one or more independently selected from the group consisting of the following compounds and pharmaceutically acceptable salts thereof:
/>
/>
wherein n is 1 to 10.
In one embodiment, the radioactive conjugate of the present invention comprises a chelator of the formula:
the chelator may be covalently attached to an antibody or antigen binding domain (e.g., h11B 6) to form an immunoconjugate or radioimmunoconjugate by reacting a compound with an azide-labeled antibody or antigen binding domain to form a 1,2, 3-triazole linker, e.g., via a click chemistry reaction, as described in WO2020/229974, or as described in PCT/IB 2021/060350.
According to the present disclosure, the chelators, radiometal complexes and radioimmunoconjugates of the invention may be produced by any method known in the art; for example, pendant aromatic/heteroaromatic groups may be attached to the macrocyclic moiety by methods known in the art, such as those illustrated and described in WO2020/229974 and PCT/IB 2021/060350.
Chemical naming
Those skilled in the art will appreciate that the structures of a compound may be named or identified using commonly accepted naming systems and symbols. By way of example, the compound may be named or identified by a common name, a system name, or a non-system name. Naming systems and symbols commonly recognized in the chemical arts include, but are not limited to, chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (IUPAC).
In general, reference to an element such as hydrogen or H is intended to include all isotopes of that element. For example, if an R group is defined to include hydrogen or H, it also includes deuterium and tritium. Containing radioactive isotopes such as tritium, C 14 、P 32 And S is 35 The compounds of (2) are thus within the scope of the present technology. Methods of inserting such markers into compounds of the present technology will be apparent to those skilled in the art based on the disclosure herein.
The term "substituted" means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that all normal valences are maintained and that the substitution results in a stable compound. When a particular group is "substituted," the group may have one or more substituents, preferably one to five substituents, more preferably one to three substituents, most preferably one to two substituents, independently selected from the list of substituents. For example, "substituted" refers to an organic group (e.g., an alkyl group) as defined below in which one or more bonds to a hydrogen atom contained therein are replaced with a bond to a non-hydrogen atom or a non-carbon atom. Substituted groups also include those wherein one or more bonds to a carbon atom or a hydrogen atom are replaced with one or more bonds to a heteroatom, including double or triple bonds. Thus, unless otherwise indicated, a substituted group is substituted with one or more substituents. In some embodiments, a substituted group is substituted with 1, 2, 3, 4, 5, or 6 substituents. Examples of substituent groups include: halogen (i.e., F, cl, br and I); a hydroxyl group; alkoxy, alkenyloxy, aryloxy, aralkoxy, heterocyclyl, heterocyclylalkyl, heterocyclyloxy and heterocyclyloxy groups; carbonyl (oxo); a carboxylate salt; an ester; a carbamate; an oxime; a hydroxylamine; an alkoxyamine; aralkoxy amines; a mercaptan; a sulfide; sulfoxide; sulfone; a sulfonyl group; pentafluorosulfanyl (i.e., SF), sulfonamide; an amine; an N-oxide; hydrazine; a hydrazide; hydrazone; an azide; an amide; urea; an amidine; guanidine; enamines; an imide; an isocyanate; isothiocyanate; cyanate ester; thiocyanate esters; an imine; a nitro group; nitrile (i.e., CN); etc. When used with reference to substituents, the term "independently" means that when there may be more than one such substituent, such substituents may be the same or different from each other.
Substituted ring groups such as substituted cycloalkyl, aryl, heterocyclyl, and heteroaryl groups also include rings and ring systems in which the bond to a hydrogen atom is replaced by a bond to a carbon atom. Thus, substituted cycloalkyl, aryl, heterocyclyl and heteroaryl groups may also be substituted with substituted or unsubstituted alkyl, alkenyl and alkynyl groups as defined below.
As used herein, cm-Cn, such as C, when used prior to a group 1 -C 11 、C 1 -C 8 Or C 1 -C 6 Refers to groups containing m to n carbon atoms.
Alkyl groups include straight and branched alkyl groups having from 1 to 12 carbon atoms, and typically from 1 to 10 carbons, or in some embodiments, from 1 to 8, 1 to 6, or 1 to 4 carbon atoms; for example, the alkyl group may contain 1 to 12 carbon atoms (C 1-12 Alkyl), or 1 to 8 carbon atoms (C 1-8 Alkyl), or 1 to 6 carbon atoms (C 1-6 Alkyl). Examples of the straight-chain alkyl group include groups such as methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched alkyl groups include, but are not limited to, isopropyl, isobutyl, sec-butyl, tert-butyl, neopentyl, isopentyl, and 2, 2-dimethylpropyl groups. The alkyl group may be substituted or unsubstituted. Representative substituted alkyl groups can be substituted one or more times with substituents such as those listed above, and include, but are not limited to, haloalkyl (e.g., trifluoromethyl), hydroxyalkyl, thioalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, alkoxyalkyl, carboxyalkyl, and the like.
Cycloalkyl groups include monocyclic, bicyclic, or tricyclic alkyl groups having 3 to 12 carbon atoms in the ring, or in some embodiments, 3 to 10, 3 to 8, or 3 to 4, 5, or 6 carbon atoms. Exemplary monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, cycloalkyl groups have 3 to 8 ring members, while in other embodiments the number of ring carbon atoms ranges from 3 to 5, 3 to 6, or 3 to 7. Bicyclic and tricyclic ring systems include bridged cycloalkyl groups and fused rings such as, but not limited to, bicyclo [2.1.1] hexane, adamantyl, decalinyl, and the like. Cycloalkyl groups may be substituted or unsubstituted. The substituted cycloalkyl groups may be substituted one or more times with non-hydrogen groups and non-carbon groups as defined above. However, substituted cycloalkyl groups also include rings substituted with straight or branched chain alkyl groups as defined above. Representative substituted cycloalkyl groups may be monosubstituted or substituted more than once, such as, but not limited to, a 2, 2-disubstituted, 2, 3-disubstituted, 2, 4-disubstituted, 2, 5-disubstituted or 2, 6-disubstituted cyclohexyl group that may be substituted with substituents such as those listed above.
A cycloalkylalkyl group is an alkyl group as defined above, wherein the hydrogen bond or carbon bond of the alkyl group is replaced by a bond to a cycloalkyl group as defined above. In some embodiments, the cycloalkylalkyl group has 4 to 16 carbon atoms, 4 to 12 carbon atoms, and typically 4 to 10 carbon atoms. The cycloalkylalkyl group may be substituted or unsubstituted. The substituted cycloalkylalkyl group may be substituted at the alkyl portion, the cycloalkyl portion, or both the alkyl portion and the cycloalkyl portion of the group. Representative substituted cycloalkylalkyl groups may be monosubstituted or substituted more than once, such as but not limited to monosubstituted, disubstituted or trisubstituted with substituents such as those listed above.
Alkenyl groups include straight and branched alkyl groups as defined above, except that there is at least one double bond between two carbon atoms. The alkenyl group has 2 to 12 carbon atoms, and typically 2 to 10 carbons, or in some embodiments, 2 to 8, 2 to 6, or 2 to 4 carbon atoms. In some embodiments, an alkenyl group may have one carbon-carbon double bond or multiple carbon-carbon double bonds, such as 2, 3, 4, or more carbon-carbon double bonds. Examples of alkenyl groups include, but are not limited to, methine, ethenyl, propenyl, butenyl, and the like. The alkenyl group may be substituted or unsubstituted. Representative substituted alkenyl groups may be monosubstituted or substituted more than once, such as but not limited to monosubstituted, disubstituted or trisubstituted with substituents such as those listed above.
Cycloalkenyl groups include cycloalkyl groups as defined above having at least one double bond between two carbon atoms. Cycloalkenyl groups may be mono-or multicyclic alkyl groups having 3 to 12, more preferably 3 to 8, carbon atoms in the ring and containing at least one double bond between the two carbon atoms. Cycloalkenyl groups may be substituted or unsubstituted. In some embodiments, cycloalkenyl groups may have one, two, or three double bonds or multiple carbon-carbon double bonds, such as 2, 3, 4, or more carbon-carbon double bonds, but do not include aromatic compounds. Cycloalkenyl groups have 3 to 14 carbon atoms, or in some embodiments, 5 to 14 carbon atoms, 5 to 10 carbon atoms, or even 5, 6, 7, or 8 carbon atoms. Examples of cycloalkenyl groups include cyclohexenyl, cyclopentenyl, cyclohexadienyl, cyclobutenyl and cyclopentadienyl.
Cycloalkenyl alkyl groups are alkyl groups as defined above wherein the hydrogen bond or carbon bond of the alkyl group is replaced by a bond to the cycloalkenyl group as defined above. Cycloalkenyl alkyl groups may be substituted or unsubstituted. The substituted cycloalkenylalkyl group may be substituted at the alkyl portion, the cycloalkenyl portion, or both the alkyl portion and the cycloalkenyl portion of the group. Representative substituted cycloalkenylalkyl groups may be substituted one or more times with substituents such as those listed above.
Alkynyl groups include straight and branched alkyl groups as defined above except that there is at least one triple bond between two carbon atoms. Alkynyl groups have 2 to 12 carbon atoms, and typically 2 to 10 carbons, or in some embodiments, 2 to 8, 2 to 6, or 2 to 4 carbon atoms. In some embodiments, the alkynyl group has one, two, or three carbon-carbon triple bonds. Examples include, but are not limited to, -c=ch, -c=cch 3 、-CH 2 C=CCH 3 、-C=CCH 2 CH(CH 2 CH 3 ) 2 Etc. Alkynyl groups may be substituted or unsubstituted. The terminal alkyne has at least one hydrogen atom bonded to a triple bond carbon atom. Representative substituted alkynyl groups may be monosubstituted or substituted more than once, such as but not limited to monosubstituted, disubstituted or trisubstituted with substituents such as those listed above. A "cyclic alkyne" or "cycloalkynyl" is a cycloalkyl ring that contains at least one triple bond between two carbon atoms. Examples of cyclic alkyne or cycloalkynyl groups include, but are not limited to, cyclooctyne, bicyclonyne (BCN), cyclooctyne Difluoride (DIFO), dibenzocyclooctyne (DIBO), keto-DIBO, diarylazedox (BARAC), dibenzoazacyclooctyne (DIBAC), dimethoxyazacyclooctyne (DIMAC), difluorobenzocyclooctyne (dibbo), monobenzocyclooctyne (MOBO), and Tetramethoxydibo (TMDIBO).
Aryl groups are cyclic aromatic hydrocarbons that contain no heteroatoms. Aryl groups herein include monocyclic, bicyclic and tricyclic ring systems. Thus, aryl groups include, but are not limited to, phenyl, azulenyl (azulenyl), cycloheptenyl (heptalenyl), biphenyl, fluorenyl, phenanthryl, anthracenyl, indenyl, indanyl, pentalenyl (pentalenyl), and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portion of the group, and in other embodiments 6 to 12 or even 6-10 carbon atoms. In some embodiments, the aryl group is phenyl or naphthyl. The aryl group may be substituted or unsubstituted. The phrase "aryl group" includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, etc.). Representative substituted aryl groups may be monosubstituted or substituted more than once. For example, monosubstituted aryl groups include, but are not limited to, 2-substituted, 3-substituted, 4-substituted, 5-substituted or 6-substituted phenyl or naphthyl groups that may be substituted with substituents such as those listed above. Aryl moieties are well known and are described, for example, in Lewis, r.j. Editions, hawley's Condensed Chemical Dictionary, 13 th edition, john Wiley & Sons, inc., new York (1997). The aryl group may be a monocyclic structure (i.e., monocyclic) or comprise a polycyclic structure (i.e., polycyclic) which is a fused ring structure. Preferably, the aryl group is a monocyclic aryl group.
An alkoxy group is a hydroxyl group (-OH) in which the bond to a hydrogen atom is replaced by a bond to a carbon atom of a substituted or unsubstituted alkyl group as defined above. Examples of straight chain alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy, and the like. Examples of branched alkoxy groups include, but are not limited to, isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy, and the like. Examples of cycloalkyloxy groups include, but are not limited to, cyclopropyloxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and the like. The alkoxy group may be substituted or unsubstituted. Representative substituted alkoxy groups may be substituted one or more times with substituents such as those listed above.
Similarly, alkylthio or thioalkoxy refers to an-SR group, wherein R is an alkyl group attached to the parent molecule through a sulfur bridge, such as-S-methyl, -S-ethyl, and the like. Representative examples of alkylthio groups include, but are not limited to, -SCH 3 、-SCH 2 CH 3 Etc.
The term "halogen" as used herein refers to bromine, chlorine, fluorine or iodine. Accordingly, the term "halo" refers to fluoro, chloro, bromo or iodo. In some embodiments, the halogen is fluorine. In other embodiments, the halogen is chlorine or bromine.
The terms "hydroxy" and "hydroxyl" are used interchangeably and refer to-OH.
The term "carboxy" refers to-COOH.
The term "cyano" refers to-CN.
The term "nitro" refers to-NO 2
The term "isothiocyanate" refers to-n=c=s.
The term "isocyanate" refers to-n=c=o.
The term "azido" refers to-N 3
The term "amino" refers to-NH 2 . The term "alkylamino" refers to an amino group in which one or both of the hydrogen atoms attached to the nitrogen is replaced with an alkyl group. The alkylamine group may be represented as-NR 2 Wherein each R is independently hydrogen or an alkyl group. For example, alkylamines include methylamine (-NHCH) 3 ) Dimethylamine (-N (CH) 3 ) 2 )、-NHCH- 2 CH 3 Etc. As used herein, the term "aminoalkyl" is intended to include both branched saturated aliphatic hydrocarbon groups and straight saturated aliphatic hydrocarbon groups substituted with one or more amino groups. Representative examples of aminoalkyl groups include, but are not limited to, -CH 2 NH 2 、-CH 2 CH 2 NH 2 and-CH 2 CH(NH 2 )CH 3
As used herein, "amide" refers to-C (O) N (R) 2 Wherein each R is independently an alkyl group or hydrogen. Examples of amides include, but are not limited to, -C (O) NH 2 、-C(O)NHCH 3 and-C (O) N (CH) 3 ) 2
The terms "hydroxyalkyl" and "hydroxyalkyl" are used interchangeably and refer to an alkyl group substituted with one or more hydroxyl groups. Alkyl groups may be branched or straight chain aliphatic hydrocarbons. Examples of hydroxyalkyl groups include, but are not limited to, hydroxymethyl (-CH) 2 OH, hydroxyethyl ]-CH 2 CH2 OH), and the like.
As used herein, the term "heterocyclyl" includes stable mono-and polycyclic hydrocarbons containing at least one heteroatom ring member, such as sulfur, oxygen or nitrogen. As used herein, the term "heteroaryl" includes stable monocyclic and polycyclic aromatic hydrocarbons containing at least one heteroatom ring member, such as sulfur, oxygen, or nitrogen. Heteroaryl groups may be monocyclic or polycyclic, for example bicyclic or tricyclic. Each ring of a heterocyclyl or heteroaryl group containing a heteroatom may contain one or two oxygen or sulfur atoms and/or one to four nitrogen atoms, provided that the total number of heteroatoms in each ring is four or less and that each ring has at least one carbon atom. A polycyclic (e.g., bicyclic or tricyclic) heteroaryl group must include at least one fully aromatic ring, but another one or more fused rings may be aromatic or non-aromatic. The heterocyclyl or heteroaryl group may be attached at any available nitrogen or carbon atom of any ring of the heterocyclyl or heteroaryl group. Preferably, the term "heteroaryl" refers to a 5-or 6-membered monocyclic group and a 9-or 10-membered bicyclic group having at least one heteroatom (O, S or N) in at least one ring, wherein the heteroatom-containing ring preferably has 1, 2 or 3 heteroatoms, more preferably 1 or 2 heteroatoms, selected from O, S and/or N. The nitrogen heteroatom of the heteroaryl group may be substituted or unsubstituted. In addition, the nitrogen and sulfur heteroatoms of the heteroaryl groups may optionally be oxidized (i.e., N→O and S (O) r Where r is 0, 1 or 2).
The term "ester" means-C (O) 2 R, wherein R is alkyl.
The term "carbamate" refers to-OC (O) NR 2 Wherein each R is independently alkyl or hydrogen.
The term "aldehyde" refers to-C (O) H.
The term "carbonate" refers to-OC (O) OR, where R is alkyl.
The term "maleimide" refers to a compound of formula H 2 C 2 (CO) 2 NH groups. The term "maleimide group" refers to a maleimide group covalently linked to another group or molecule. Preferably, the maleimide group is N-linked, for example:
the term "acyl halide" refers to-C (O) X, wherein X is halo (e.g., br, cl). Exemplary acid halides include acid chloride (-C (O) Cl) and acid bromide (-C (O) Br).
According to the convention used in the art:
as used in the formulae herein to describe bonds as attachment points of moieties, functional groups or substituents to a core, parent or backbone structure, such as an antigen binding domain of the invention.
When any variable occurs more than once in any component or formula of a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-3R groups, then the group may optionally be substituted with up to three R groups, and at each occurrence R is independently selected from the definition of R.
When the bond to a substituent is shown as intersecting a bond connecting two atoms in a ring, then such substituent may be bound to any atom on the ring.
In certain embodiments, the radioactive conjugate is 225 Ac-DOTA-h11B6, also known as actinium 225-1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid-h 11B6. As used herein, 225 Ac-DOTA-h11B6 is a polypeptide comprising a chelate to DOTA 225 A radioactive conjugate of Ac, wherein the DOTA is conjugated to h11B6, optionally via a linker. As used herein, 111 In-DOTA-h11B6 is a composition comprising a chelate to DOTA 111 A radioactive conjugate of In, wherein the DOTA is conjugated to h11B6, optionally via a linker.
In certain embodiments, the radioactive conjugate may be represented by the following compounds or variants thereof:
in certain embodiments, the radioactive conjugate is 225 Ac-TOPA-h11B6. As used herein, 225 Ac-TOPA-h11B6 is a peptide comprising chelate to TOPA 225 A radioactive conjugate of Ac, wherein the TOPA is conjugated to h11B6, optionally via a linker. In one embodiment of the present invention, in one embodiment, 225 Ac-TOPA-h11B6 radioactive conjugates can be represented by the following compounds or variants thereof, which can also be referred to as TOPA- [ C7]Phenylthiourea-h 11B6 antibody conjugate:
/>
in the TOPA- [ C7] -phenylthiourea-h 11B6 antibody conjugate described above, the structure does not show a lysine residue of h11B6 attached to the phenylthiourea moiety, as shown in FIGS. 6B and 6C.
Pharmaceutical compositions and methods of use
Embodiments of the present invention provide methods of treating cancer in a patient comprising administering to the patient a therapeutically effective amount of a radioconjugate. According to one embodiment, the method comprises administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radio conjugate and one or more pharmaceutically acceptable excipients.
Embodiments of the invention are particularly useful in treating patients who have been diagnosed with prostate cancer; for example, patients with advanced prostate cancer. According to one embodiment, the cancer is non-restricted prostate cancer. According to another embodiment, the cancer is metastatic prostate cancer. According to another embodiment, the cancer is Castration Resistant Prostate Cancer (CRPC). According to another embodiment, the cancer is metastatic castration-resistant prostate cancer (mCRPC). According to another embodiment, the cancer is mCRPC with an adenocarcinoma. According to particular embodiments, the patient's testosterone castration level is about 50ng/dL or less. According to additional embodiments, the patient has been previously exposed to at least one Androgen Receptor (AR) targeted therapy; for example, abiraterone acetate, enzalutamide, apatamide, darunamine, or a combination of any of the foregoing. According to additional embodiments, the patient has previously undergone chemotherapy; for example, chemotherapy includes administration of taxanes. According to another embodiment, the patient has previously undergone orchiectomy or medical castration. According to another embodiment, the patient is receiving androgen deprivation therapy with a gonadotropin releasing hormone (GnRH) agonist or antagonist.
According to embodiments of the methods of treatment described herein, the radioactive conjugate administered to a patient comprises at least one radioactive metal complex conjugated to an antibody or antigen-binding fragment having binding specificity for hK 2. Preferably, the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2. In a preferred embodiment, the radioactive metal complex comprises 225 Ac。
According to one embodiment, the radioactive metal in the pharmaceutical composition is 225 Ac and provides targeted radioactivity of about 50 to about 350 μci per dose of the pharmaceutical composition. According to additional embodiments, the radiometal in the pharmaceutical composition provides a target of about 50 to about 300, or about 50 to about 250, or about 50 to about 240, or about 50 to about 230, or about 50 to about 220, or about 50 to about 210, or about 50 to about 200, or about 50 to about 175, or about 50 to about 150, or about 50 to about 125, or about 50 to about 100, or about 100 to about 300, or about 100 to about 250, or about 100 to about 240, or about 100 to about 230, or about 100 to about 220, or about 100 to about 210, or about 100 to about 200, or about 100 to about 175, or about 150 to about 150, or about 150 to about 300 of the pharmaceutical composition.
According to one embodiment, the radioactive metal in the pharmaceutical composition is 225 Ac and provides about 50 μci to about 500 μ per dose of the pharmaceutical compositionTargeted radioactivity of Ci. According to additional embodiments, the radiometal in the pharmaceutical composition provides targeted radioactivity per dose of the pharmaceutical composition from about 50 μci to about 450 μci, or from about 50 μci to about 400 μci, or from about 50 μci to about 350 μci, or from about 100 μci to about 500 μci, or from about 100 μci to about 450 μci, or from about 100 μci to about 400 μci, or from about 100 μci to about 350 μci, or from about 150 μci to about 500 μci, or from about 150 μci to about 450 μci, or from about 150 μci to about 400 μci, or from about 150 μci to about 350 μci, or from about 200 μci to about 500 μci, or from about 200 μci to about 450 μci, or from about 200 μci to about 400 μci.
According to one embodiment, the radioactive metal in the pharmaceutical composition is 225 Ac, and provides a targeted specific activity of about 50 μci to about 350 μci per about 2mg total antibodies in the pharmaceutical composition. According to additional embodiments, the radioactive metal in the pharmaceutical composition provides a specific activity of about 50 to about 300, or about 50 to about 250, or about 50 to about 240, or about 50 to about 230, or about 50 to about 220, or about 50 to about 210, or about 50 to about 200, or about 50 to about 175, or about 50 to about 150, or about 50 to about 125, or about 50 to about 100, or about 100 to about 300, or about 100 to about 250, or about 100 to about 240, or about 100 to about 230, or about 100 to about 220, or about 100 to about 210, or about 100 to about 200, or about 100 to about 175, or about 150 to about 150, or about 150 to about 250 per about 2mg of total antibody in the pharmaceutical composition. According to additional embodiments, the radioactive metal in the pharmaceutical composition provides a targeted specific activity of about 50 μci, or about 100 μci, or about 150 μci, or about 175 μci, or about 200 μci, or about 225 μci, or about 250 μci, or about 275 μci, or about 300 μci per about 2mg of total antibody in the pharmaceutical composition. For example, it should be understood that 300 μCi per 2mg of antibody is equivalent to 150 μCi/mg of antibody, 200 μCi per 2mg of antibody is equivalent to 100 μCi/mg of antibody, and so forth.
According to another embodiment, the radiometal in the pharmaceutical compositionIs that 225 Ac, and provides a targeted specific activity of between about 50 μci to about 350 μci per about 2mg and about 10mg of total antibody in the pharmaceutical composition (e.g., per about 2mg, or about 4mg, or about 6mg, or about 8mg, or about 10mg of total antibody in the pharmaceutical composition). According to additional embodiments, the radioactive metal in the pharmaceutical composition provides between about 2mg and about 10mg of total antibody per about 2mg of the pharmaceutical composition (e.g., about 2mg, or about 4mg, or about 6mg, or about 8mg, or about 10mg of total antibodies in the pharmaceutical composition) about 50 to about 300, or about 50 to about 250, or about 50 to about 240, or about 50 to about 230, or about 50 to about 220, or about 50 to about 210, or about 50 to about 200, or about 50 to about 175, or about 50 to about 150, or about 50 to about 125, or about 50 to about 100, or about 100 to about 300, or about 100 to about 230, or about 100 to about 220, or about 100 to about 210, or about 100 to about 200, or about 100 to about 175, or about 150 to about 250, or about 150 to about 100 Ci. According to additional embodiments, the radioactive metal in the pharmaceutical composition provides a targeted specific activity of between about 2mg and about 10mg of total antibody in the pharmaceutical composition (e.g., about 50 μci, or about 100 μci, or about 150 μci, or about 175 μci, or about 200 μci, or about 225 μci, or about 250 μci, or about 275 μci, or about 300 μci, or about 350 μci of total antibody in the pharmaceutical composition).
According to another embodiment, the radioactive metal in the pharmaceutical composition is 225 Ac, and provides a targeted specific activity of about 50 to about 500 μci per about 2mg and about 10mg of total antibody in the pharmaceutical composition (e.g., per about 2mg, or about 4mg, or about 6mg, or about 8mg, or about 10mg of total antibody in the pharmaceutical composition). According to additional embodiments, the radioactive metal in the pharmaceutical composition provides between about 2mg and about 10mg of total antibody per about 2mg of the pharmaceutical composition (e.g., about 4mg, or about 6mg, per about 2mg, or about 4mg, or about 6mg of total antibody per about,Or about 8mg, or about 10mg total antibody) from about 50 μci to about 450 μci, or from about 50 μci to about 400 μci, or from about 50 μci to about 350 μci, or from about 100 μci to about 500 μci, or from about 100 μci to about 450 μci, or from about 100 μci to about 400 μci, or from about 100 μci to about 350 μci, or from about 150 μci to about 500 μci, or from about 150 μci to about 450 μci, or from about 150 μci to about 400 μci, or from about 150 μci to about 350, or from about 200 μci to about 500 μci, or from about 200 μci to about 450 μci, or from about 200 μci to about 350 μci. According to additional embodiments, the radioactive metal in the pharmaceutical composition provides a targeted specific activity of between about 350 μci, or about 375 μci, or about 400 μci, or about 425 μci, or about 450 μci, or about 475 μci, or about 500 μci per about 2mg and about 10mg of total antibodies in the pharmaceutical composition (e.g., per about 2mg, or about 4mg, or about 8mg, or about 10mg of total antibodies in the pharmaceutical composition).
According to one embodiment, the radioactive metal in the pharmaceutical composition is 225 Ac, and the targeted radioactivity concentration of the pharmaceutical composition is about 1 μCi/mL to about 100 μCi/mL, or about 5 μCi/mL to about 75 μCi/mL, or about 10 μCi/mL to about 60 μCi/mL, or about 12.5 μCi/mL to about 50 μCi/mL, or about 12.5 μCi/mL, or about 25 μCi/mL, or about 37.5 μCi/mL, or about 50 μCi/mL.
As used herein, "time of administration" refers to the time when a patient is administered a dose of a pharmaceutical composition comprising a radioactive conjugate (e.g., whether as a single administration or multiple administrations in more than one sub-dose). Due to 225 Decay of Ac, in pharmaceutical compositions consisting of 225 The amount of radioactivity provided by Ac decreases from the time of preparation to the time of administration, i.e., from the time of preparation 225 The approximate time that Ac chelates to the conjugate intermediate to form the radioactive conjugate is to the time that the radioactive conjugate is administered to the patient. According to one example, if made from a pharmaceutical composition 225 Ac provided radioactivity at 225 The approximate time for Ac to chelate to the conjugate intermediate to form a radioactive conjugate is about 264 μci (e.g., after the radioactive conjugate is formed and purified), then the radioactivity after about 96 hours at the time of administration may be about 200 μci. Decay and process for producing the same Thus at any given time 225 The amount of Ac may be based on the initial amount of activity measured at time zero, the amount of time elapsed, and 225 the half-life of Ac was calculated.
As used herein, "targeted" specific activity or "targeted" radioactivity or "targeted" concentration of radioactivity refers to the amount of specific activity or radioactivity or concentration of radioactivity, respectively, calculated to be present in a dose of a pharmaceutical composition at the desired time of administration to a patient, e.g., based on the amount of radioactive metal present in the composition at the time of preparation and the desired amount of time (and concomitant decay of radioactive metal) between preparation and administration to the patient. It will be appreciated that the actual specific activity or radioactivity or concentration of radioactivity at the time of administration may be slightly different from the targeted specific activity or concentration of radioactivity or radioactivity, respectively (e.g., where the actual administration time of the patient is slightly different from the intended administration time).
According to a particular embodiment, the pharmaceutical composition further comprises a non-radiolabeled antibody. For example, a composition comprising a non-radiolabeled antibody may be combined with a composition comprising a radioactive conjugate to dilute the radioactive conjugate composition to a desired dose of radioactivity. As used herein, the term "non-radiolabeled antibody" refers to an antibody or antibody-chelator complex that is not conjugated to a radioactive metal. According to a particular embodiment, the non-radiolabeled antibody present in the composition is a conjugate intermediate, such as DOTA-mAb (e.g., DOTA-h11B 6). Preferably, the non-radiolabeled antibody comprises the same antibody as the radiolabeled conjugate contained in the composition; for example, the pharmaceutical composition may comprise an amount of 225 Ac-DOTA-h11B6 and an amount of DOTA-h11B6. Alternatively, the pharmaceutical composition may comprise an amount of 225 Ac-TOPA-h11B6 and an amount of TOPA-h11B6. As used herein, "total antibody" refers to the total amount of antibody in a pharmaceutical composition; for example, the total antibodies can include (a) an amount of antibody conjugated to a radioactive metal complex and (b) an amount of non-radiolabeled antibody (such as a conjugate intermediate). Targeting total antibody refers to calculating the antibodies present in a dose of a pharmaceutical composition at the time of intended administration to a patientIs a combination of the amounts of (a) and (b). According to certain embodiments, the total amount of antibodies (radiolabeled and non-radiolabeled) in the composition is no more than about 10mg, or about 9mg, or about 8mg, or about 7mg, or about 6mg, or about 5mg, or about 4mg, or about 3mg, or about 2mg.
According to certain embodiments, a method of preparing a pharmaceutical composition of the invention comprises combining a first intermediate composition and a second intermediate composition to form the pharmaceutical composition, wherein: the first intermediate composition comprises a radioactive conjugate and the second intermediate composition comprises a conjugate intermediate and does not comprise any radioactive conjugate. According to certain embodiments, the first intermediate composition and the second intermediate composition comprise the same pharmaceutically acceptable excipient.
According to a specific embodiment of the present invention, the pharmaceutical composition comprises about 0.1mg to about 5mg of the total antibody, or about 0.1mg to about 4mg of the total antibody, or about 0.1mg to about 3mg of the total antibody, or about 0.1mg to about 4mg of the total antibody, about 0.1mg to about 3mg of the total antibody, about 0.1mg to about 2mg of the total antibody, or about 0.5mg to about 5mg of the total antibody, or about 0.5mg to about 4mg of the total antibody, or about 0.5mg to about 3mg of the total antibody, or about 0.5mg to about 4mg of the total antibody, or about 1mg to about 10mg of the total antibody, or about 1mg to about 7mg of the total antibody, or about 1mg to about 5mg of the total antibody, or about 1mg to about 4mg of the total antibody or about 1mg to about 3mg of the total antibody, or about 1.5mg to about 2.5mg of the total antibody, or about 1.1mg to about 1.2mg of the total antibody, or about 1.3mg of the total antibody, or about 1.4mg of the total antibody, or about 1.5mg of the total antibody, or about 1.6mg of the total antibody, or about 1.7mg of the total antibody, or about 1.8mg of the total antibody, or about 1.9mg of the total antibody, or about 2mg of the total antibody, or about 2.1mg of the total antibody, or about 2.2mg of the total antibody, or about 2.3mg of the total antibody, or about 2.4mg of the total antibody, or about 2.5mg of the total antibody, or about 2.6mg of the total antibody, or about 2.7mg of the total antibody, or about 2.8mg of the total antibody, or about 2.9mg of the total antibody.
According to particular embodiments, the dose of the pharmaceutical composition has a volume of about 1mL to about 20mL, or about 1mL to about 10mL, or about 2mL to about 6mL, or about 3mL to about 5mL, or about 4 mL. According to one embodiment, the dose of the pharmaceutical composition comprises about 2mg of total antibody per about 4mL dose (i.e., about 1mg of total antibody per about 2mL dose). As described herein, the dose may be administered as a plurality of sub-doses; for example, an 8mL dose may be administered as two 4mL sub-doses. In one embodiment, two 4mL sub-doses, each containing 2mg of antibody, are administered to the subject, for a total of 4mg of antibody per 8mL dose.
According to particular embodiments, the pharmaceutical composition comprises total antibodies in an amount of about 0.01mg/mL to 5.0mg/mL, or about 0.01mg/mL to 4.0mg/mL, or about 0.01mg/mL to 3.0mg/mL, about 0.01mg/mL to 2.0mg/mL, or about 0.01mg/mL to 1.0mg/mL, about 0.1mg/mL to 5.0mg/mL, or about 0.1mg/mL to 4.0mg/mL, or about 0.1mg/mL to 3.0mg/mL, about 0.1mg/mL to 2.0mg/mL, or about 0.1mg/mL to 1.0mg/mL, about 0.3mg/mL to 0.7mg/mL, or about 0.4mg/mL to 0.6mg/mL, or about 0.5 mg/mL.
According to one embodiment, the targeted total antibody concentration in a vial containing a dose of the pharmaceutical composition is about 0.5mg/ml±0.1mg/mL; for example, in the presence of 225 Vials of Ac-DOTA-h11B6 and DOTA-h11B 6. In one embodiment, if a dose contains about 4mL of the pharmaceutical composition, the targeted total antibody concentration in the dose is about 0.5mg/mL, the targeted radioactivity of the dose is about 50 μci, and the targeted radioactivity concentration is about 12.5 μci/mL (e.g., 12.5 μci/ml±10%). In another embodiment, if a dose contains about 4mL of the pharmaceutical composition, the targeted total antibody concentration in the dose is about 0.5mg/mL, the targeted radioactivity of the dose is about 100 μci, and the targeted radioactivity concentration is about 25 μci/mL (e.g., 25 μci/ml±10%). In another embodiment, if a dose contains about 4mL of the pharmaceutical composition, the targeted total antibody concentration in the dose is about 0.5mg/mL, the targeted radioactivity of the dose is about 150 μci, and the targeted radioactivity concentration is about 37.5 μci/mL (e.g., 37.5 μci/ml±10%). In another embodiment, if a dose contains about 4mL of the pharmaceutical composition, the targeted total antibody concentration in the dose is about 0.5mg/mL (about 2mg total antibody), the targeted radioactivity of the dose is about 200 μci, and the targeted radioactivity concentration is about 50 μci/mL (e.g., 50 μci/ml±10%). In another embodiment, if a dose contains about 8mL of the pharmaceutical composition The total targeted antibody concentration in this dose was about 0.5mg/mL (about 4mg total antibody), the targeted radioactivity in this dose was about 300 μci, and the targeted radioactivity concentration was about 37.5 μci/mL (e.g., 37.5 μci/ml±10%).
According to other embodiments, the total antibody is present in the pharmaceutical composition at a concentration of about 0.1mg/mL to about 1mg/mL. In some embodiments of the present invention, in some embodiments, about 0.1mg/mL of total antibody in the pharmaceutical composition is about 0.1mg/mL to about 0.9mg/mL, about 0.1mg/mL to about 0.8mg/mL, about 0.1mg/mL to about 0.7mg/mL, about 0.1mg/mL to about 0.6mg/mL, about 0.1mg/mL to about 0.5mg/mL, about 0.1mg/mL to about 0.4mg/mL, about 0.1mg/mL to about 0.3mg/mL, about 0.1mg/mL to about 0.2mg/mL, about 0.2mg/mL to about 0.1mg/mL, about 0.2mg/mL to about 0.8mg/mL, about 0.2mg/mL to about 0.2mg/mL, about 0.5mg/mL to about 0.5mg/mL, about 0.0.0.5 mg/mg to about 0.3 mg/about 0.5mg/mL, about 0.0.5 mg/about 0.5mg to about 0.3mg/mL, about 0.0.5 mg/mg to about 0.3 mg/about 0.5mg/mL, about 0.0.4 mg/about 0.0 mg/mg to about 0.3 mg/about 0.5 mg/mg to about 0.3 mg/mg, about 0.6mg/mL to about 0.8mg/mL, about 0.6mg/mL to about 0.7mg/mL, about 0.7mg/mL to about 1mg/mL, about 0.7mg/mL to about 0.9mg/mL, about 0.7mg/mL to about 0.8mg/mL, about 0.8mg/mL to about 1mg/mL, about 0.8mg/mL to about 0.9mg/mL, or about 0.9mg/mL to about 1mg/mL. In other embodiments, the pharmaceutical composition contains about 0.5mg/mL total antibody. In other embodiments, the pharmaceutical composition contains a total of about 0.1mg/mL to about 1mg/mL 225 Ac-DOTA-h11B6 and DOTA-h11B6. In other embodiments, the pharmaceutical composition contains a total of about 0.5mg/mL 225 Ac-DOTA-h11B6 and DOTA-h11B6. In other embodiments, the pharmaceutical composition contains a total of about 0.1mg/mL to about 1mg/mL 225 Ac-TOPA-h11B6 and TOPA-h11B6. In which it is arrangedIn other embodiments, the pharmaceutical composition contains a total of about 0.5mg/mL 225 Ac-TOPA-h11B6 and TOPA-h11B6.
The pharmaceutical compositions of the present invention may be administered via any suitable route known to those skilled in the art. For example, the composition may be administered parenterally. Non-limiting examples of routes of administration include Intravenous (IV), intramuscular, or subcutaneous, or they may be administered by infusion techniques. In certain aspects, the methods of treatment herein comprise intravenous injection of a pharmaceutical composition.
According to one embodiment, the pharmaceutical composition of the invention is provided as a disposable sterile injectable solution. The composition is preferably cooled in a sealed vial until injection; for example, in a cyclic olefin polymer vial closed with a latex-free stopper and an aluminum seal.
The pharmaceutical compositions of the present invention may be administered to a patient by a health care professional. In some embodiments, the pharmaceutical composition is administered once every about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks, or about 16 weeks. In some embodiments, the pharmaceutical composition is administered once every about 4 weeks to about 12 weeks, about 4 weeks to about 10 weeks, about 4 weeks to about 8 weeks, about 4 weeks to about 6 weeks, about 6 weeks to about 12 weeks, about 6 weeks to about 10 weeks, about 6 weeks to about 8 weeks, about 8 weeks to about 12 weeks, about 8 weeks to about 10 weeks, about 10 weeks to about 12 weeks. In certain aspects, the pharmaceutical composition is administered to the patient once every about 4 weeks. In certain aspects, the pharmaceutical composition is administered to the patient once every about 6 weeks. In other aspects, the pharmaceutical composition is administered to the patient once every about 8 weeks. In certain aspects, the pharmaceutical composition is administered to the patient once every about 10 weeks. In other aspects, the pharmaceutical composition is administered to the patient once every about 12 weeks. According to certain embodiments, the patient is administered from about 2 to about 12 doses, or from about 2 to about 10 doses, or from about 2 to about 8 doses, or from about 2 to about 6 doses, or from about 2 to about 4 doses. According to one embodiment, the patient's dosing regimen comprises administration of at least 2 doses, or at least 3 doses, or at least 4 doses, or at least 5 doses, or at least 6 doses, wherein one dose is administered every 8 weeks. According to one embodiment, the patient's dosing regimen comprises administration of 4 total doses, with one dose administered every 8 weeks. Alternatively, the patient's dosing regimen comprises administering 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 total doses, with one dose administered every 8 weeks. In still further embodiments, the patient's dosing regimen may continue such that it includes more than 12 total doses.
The dose of the pharmaceutical composition of the invention may be administered to a patient by a single administration or by administering the dose in multiple administrations of more than one sub-dose (e.g. by administering the dose in multiple subdivisions of the dose). Alternatively, the dose may be provided as a continuous infusion over an extended period of time.
It will be appreciated by those skilled in the art that the pharmaceutical compositions of the present invention may be administered alone or in combination with one or more additional therapeutic or imaging agents or methods as determined by the attending physician. The pharmaceutical compositions of the present invention may be administered to a patient prior to or concurrently with other therapeutic methods for treating prostate cancer.
Preferably, the pharmaceutical composition of the invention is in the form of a sterile aqueous solution which may contain other substances to render the solution isotonic with blood and/or to provide a suitable pH. In some embodiments, the aqueous solution has a pH of about 4 to about 7, about 4.5 to about 6.5, about 5 to about 6, or about 5.5. In other embodiments, the aqueous solution has a pH of about 5, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, or about 6. In other embodiments, the aqueous solution has a pH of about 5.5.
As described herein, due to 225 Decay of Ac in a dose of pharmaceutical composition 225 The amount of radioactivity provided by Ac was measured from the time of preparation (from 225 The approximate time that Ac chelates to the conjugate intermediate to form the radioactive conjugate) to the time that the dose is administered to the patient. Preferably, a sufficient amount is used during the preparation 225 Ac-labeled radio conjugates to account for estimated occurrence between about chelation time and time of administration to patients 225 Reduction of Ac specific activity.It is also preferred to limit the formation of radioactive conjugates (by chelation 225 Ac) and the amount of time between administration of the dose to the patient.
According to particular embodiments, the pharmaceutical composition of the invention is administered to the patient within about 7 days (168 hours) from the time the radiometal is chelated to the conjugate intermediate to form the radioconjugate, or within about 144 hours, or within about 120 hours, or within about 96 hours, or within about 72 hours, or within about 48 hours, or within about 24 hours from the time the radiometal is chelated to the conjugate intermediate to form the radioconjugate. According to particular embodiments, the methods of the invention comprise administering the pharmaceutical composition to the patient (i.e., the time at which administration occurs) within about 120 hours or less from the time the radiometal is chelated to the conjugate intermediate to form the radioconjugate. According to particular embodiments, the methods of the invention comprise administering the pharmaceutical composition to the patient (i.e., the time at which administration occurs) within about 96 hours or less from the time the radiometal is chelated to the conjugate intermediate to form the radioconjugate. According to particular embodiments, the methods of the invention comprise administering the pharmaceutical composition to the patient (i.e., the time at which administration occurs) within about 72 hours or less from the time the radiometal is chelated to the conjugate intermediate to form the radioconjugate.
According to a particular embodiment, the radioactive conjugate of the invention is administered in admixture with one or more pharmaceutically acceptable excipients. The terms "pharmaceutical composition" and "pharmaceutical formulation" are used interchangeably throughout the disclosure. The pharmaceutical compositions may be prepared using techniques known in the art. In certain embodiments, the pharmaceutical composition has sufficient storage stability and is suitable for administration to humans.
Excipients may be selected by those skilled in the art and may take a variety of forms depending on the route of administration desired. For example, for parenteral administration, the excipients may include sterile water, and other ingredients may be added to increase the solubility and preservability of the composition. Injectable suspensions or solutions may also be prepared by including aqueous carriers and/or appropriate additives such as solubilizers and preservatives.
In some embodiments, the excipient comprises a buffer, which is an aqueous solution preferably containing an acid-base mixture, in order to stabilize the pH of the solution. Examples of buffers include, but are not limited to Trizma, bicine, genistein, MOPS, MOPSO, MOBS, tris, hepes, HEPBS, MES, phosphate, carbonate, acetate, citrate, glycolate, lactate, borate, ACES, ADA, tartrate, AMP, AMPD, AMPSO, BES, CABS, dimethylarsinate, CHES, DIPSO, EPPS, ethanolamine, glycine, HEPPSO, imidazole lactic acid, PIPES, SSC, SSPE, POPSO, TAPS, TABS, TAPSO, or TES. In some embodiments, the buffer is Trizma. In some embodiments, the buffer is Bicine. In other embodiments, the buffer is genistein. In other embodiments, the buffer is MOPS. In other embodiments, the buffer is MOPSO. In other embodiments, the buffer is MOBS. In other embodiments, the buffer is Tris. In other embodiments, the buffer is Hepes. In other embodiments, the buffer is HEPBS. In other embodiments, the buffer is MES. In other embodiments, the buffer is phosphate. In other embodiments, the buffer is a carbonate salt. In other embodiments, the buffer is acetate. In other embodiments, the buffer is citrate. In other embodiments, the buffer is glycolate. In other embodiments, the buffer is lactate. In other embodiments, the buffer is a borate. In other embodiments, the buffer is ACES. In other embodiments, the buffer is ADA. In other embodiments, the buffer is tartrate. In other embodiments, the buffer is AMP. In other embodiments, the buffer is AMPD. In other embodiments, the buffer is AMPSO. In other embodiments, the buffer is BES. In other embodiments, the buffer is CABS. In other embodiments, the buffer is dimethylarsinate. In other embodiments, the buffer is CHES. In other embodiments, the buffer is DIPSO. In other embodiments, the buffer is EPPS. In other embodiments, the buffer is ethanolamine. In other embodiments, the buffer is glycine. In other embodiments, the buffer is HEPPSO. In other embodiments, the buffer is imidazole. In other embodiments, the buffer is imidazolactic acid. In other embodiments, the buffer is PIPES. In other embodiments, the buffer is SSC. In other embodiments, the buffer is SSPE. In other embodiments, the buffer is POPSO. In other embodiments, the buffer is TAPS. In other embodiments, the buffer is TABS. In other embodiments, the buffer is TAPSO. In other embodiments, the buffer is TES. The buffer is desirably provided in a concentration to achieve the desired pH. In some aspects, the concentration of the buffer is about 10mM to about 50mM. In other aspects, the pH of the buffer is about 20mM to about 50mM, about 25mM to about 50mM, about 30mM to about 50mM, about 35mM to about 50mM, about 40mM to about 50mM, about 45mM to about 50mM, about 20mM to about 45mM, about 25mM to about 45mM, about 30mM to about 45mM, about 35mM to about 45mM, about 40mM to about 45mM, about 20mM to about 40mM, about 25mM to about 40mM, about 30mM to about 40mM, about 35mM to about 40mM, about 20mM to about 35mM, about 25mM to about 35mM, about 30mM to about 35mM, about 20mM to about 30mM, about 25mM to about 30mM, about 20mM to about 25mM. In other aspects, the buffer is at a concentration of about 24mM to about 28mM, about 25mM to about 27mM, about 26mM to about 28mM, or about 26mM to about 27mM. In other aspects, the buffer is at a concentration of about 25mM. In other aspects, the buffer is at a concentration of about 26.75mM. In one embodiment, the buffer comprises acetate.
In additional embodiments, the excipient comprises a diluent. The term "diluent" refers to an aqueous or non-aqueous solution used to dilute the pharmaceutical composition. For example, the diluent may include one or more of saline, water, polyethylene glycol, propylene glycol, ethanol, or an oil (such as safflower oil, corn oil, peanut oil, cottonseed oil, or sesame oil). In certain embodiments, the diluent is water. Additional non-limiting examples of diluents include sterile water, saline of different concentrations (NS/0.9%, 1/2 NS/0.45%), dextrose of different concentrations (D5W, D10W), or dextrose + saline (1/2 NSD 5W).
The pharmaceutical composition may be subjected to conventional pharmaceutical procedures such as sterilization and/or may contain conventional adjuvants. The pharmaceutical compositions may also contain aqueous and non-aqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats and/or solutes which render the formulation isotonic with the blood of the intended recipient.
In other embodiments, the excipient may comprise one or more of a binder, carbohydrate, coating agent, colorant, disintegrant, dispersing agent, emulsifier, filler, flavoring agent, granulating agent, lipid, lubricant, mineral, polymer, preservative, radioprotectant, solubilizing agent, stabilizer, suspending agent, sweetener, thickener, wetting agent, or a combination thereof.
According to a particular embodiment, the excipient comprises at least one radioprotectant. In certain embodiments, the pharmaceutical composition comprises: a radio conjugate and one or more pharmaceutically acceptable excipients, wherein: the radioconjugate comprises at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2 (e.g., h11B6 or a variant thereof), and the one or more pharmaceutically acceptable excipients comprise one or more radioprotective agents.
Examples of radioprotectants include, but are not limited to, sodium ascorbate, gentisic acid or combinations thereof. According to one embodiment, the composition comprises sodium ascorbate. According to an alternative embodiment, the composition comprises gentisic acid.
In other embodiments, the excipient comprises one or more surfactants. Non-limiting examples of surfactants include polysorbates and poloxamers such as polysorbate 20, polysorbate 80, and poloxamer 188. According to one embodiment, the excipient comprises polysorbate 20. In other embodiments, the excipient comprises sodium ascorbate, polysorbate 20, or a combination thereof. In some embodiments, the pharmaceutical composition contains a radioactive conjugate and sodium ascorbate. In other embodiments, the pharmaceutical composition contains polysorbate 20. In other embodiments, the pharmaceutical composition contains sodium ascorbate and polysorbate 20. In some embodiments, the pharmaceutical composition contains a radioactive conjugate and gentisic acid. In other embodiments, the pharmaceutical composition comprises gentisic acid and polysorbate 20.
The amount of excipient is selected based upon the desired route of administration, the patient, and the particular excipient in the pharmaceutical composition. In a preferred embodiment, the amount of sodium ascorbate present in the pharmaceutical composition inhibits degradation of the radioactive conjugate. Ideally, sodium ascorbate inhibits degradation of the radioactive conjugate as compared to a composition that does not contain sodium ascorbate, as measured, for example, by spectroscopic methods such as high performance liquid chromatography, nuclear magnetic resonance, mass spectrometry, or elemental analysis. In other embodiments, the pharmaceutical composition contains from about 0.05w/v% to about 5.0w/v% sodium ascorbate and/or gentisic acid. In other embodiments, the pharmaceutical composition comprises about 0.1w/v% to about 5w/v%, about 0.1w/v% to about 4w/v%, about 0.1w/v% to about 3w/v%, about 0.1w/v% to about 2w/v%, about 0.1w/v% to about 1w/v%, about 0.2w/v% to about 1w/v%, about 0.3w/v% to about 1w/v%, about 0.4w/v% to about 1w/v%, about 0.5w/v% to about 1w/v%, about 0.6w/v% to about 1w/v%, about 0.9w/v% to about 1w/v%, about 0.1w/v% to about 0.9w/v%, about 0.2w/v% to about 1w/v%, about 0.3w/v% to about 0.5w/v%, about 0.5w/v% to about 1w/v%, about 1w/v% to about 1w/v%, about 0.6w/v% to about 1w/v%, about 0.7w/v% to about 1w/v%, about 1w/v% to about 1w/v, about 0.5w/v% to about 0.6w/v%, about 0.1w/v% to about 0.5w/v%, about 0.2w/v% to about 0.5w/v%, about 0.3w/v% to about 0.5w/v%, about 0.4w/v% to about 0.5w/v%, about 0.1w/v% to about 0.4w/v%, about 0.2w/v% to about 0.4w/v%, about 0.3w/v% to about 0.4w/v%, about 0.1w/v% to about 0.3w/v%, about 0.2w/v% to about 0.3w/v%, or about 0.1w/v% to about 0.2w/v% sodium ascorbate. In other embodiments, the pharmaceutical composition contains about 0.lw/v%, about 0.2w/v%, about 0.3w/v%, about 0.4w/v%, about 0.5w/v%, about 0.6w/v%, about 0.7w/v%, about 0.8w/v%, about 0.9w/v%, or about 1w/v% sodium ascorbate. In other embodiments, the pharmaceutical composition contains about 0.5w/v% sodium ascorbate.
In other embodiments, the pharmaceutical composition contains about 0.005 to about 0.15, or about 0.005 to about 0.12, or about 0.005 to about 0.1, or about 0.005 to about 0.08, or about 0.005 to about 0.06, or about 0.005 to about 0.12, or about 0.005 to about 0.04w/v% polysorbate 20. In certain aspects, the pharmaceutical composition comprises about 0.01w/v% to about 0.12w/v%, about 0.02w/v% to about 0.12w/v%, about 0.03w/v% to about 0.12w/v%, about 0.05w/v% to about 0.12w/v%, about 0.06w/v% to about 0.12w/v%, about 0.07w/v% to about 0.12w/v%, about 0.08w/v% to about 0.12w/v%, about 0.09w/v% to about 0.12w/v%, about 0.01w/v% to about 0.1w/v%, about 0.02w/v% to about 0.1w/v%, about 0.04w/v% to about 0.1 w% about 0.09w/v%, about 0.06 w% to about 0.09 w% to about 0.09 w% 0.12 w% to about 0.09 w% about 0.0.08 w% to about 0.0.0.04 w% to about 0.12 w% about 0.12 w% to about 0.12 w% of, about 0.07w/v% to about 0.08w/v%, about 0.01w/v% to about 0.07w/v%, about 0.02w/v% to about 0.07w/v%, about 0.03w/v% to about 0.06w/v%, about 0.04w/v% to about 0.06w/v%, about 0.05w/v% to about 0.06w/v%, about 0.01w/v% to about 0.05w/v%, about 0.02w/v% to about 0.05w/v%, about 0.03w/v% to about 0.05w/v%, about 0.04w/v% to about 0.04w/v%, about 0.03w/v% to about 0.04w/v%, about 0.01w/v% to about 0.01w/v%, about 0.03w/v% to about 0.02w/v%, or about 0.02w/v% to about 0.02w/v% of the polysorbate. In other aspects, the pharmaceutical composition contains about 0.01w/v%, about 0.02w/v%, about 0.03w/v%, about 0.04w/v%, about 0.05w/v%, about 0.06w/v%, about 0.07w/v%, about 0.08w/v%, about 0.09w/v%, about 0.1w/v%, about 0.11w/v%, about 0.12w/v%, about 0.13w/v%, about 0.14w/v%, or about 0.15w/v% polysorbate 20. In other aspects, the pharmaceutical composition contains about 0.04w/v% polysorbate 20.
According to certain embodiments, the pharmaceutical composition does not contain any preservative.
According to certain embodiments, the pharmaceutical composition does not contain any sucrose; in particular, when the radioactive metal is 225 Ac, e.g. when the radioactive conjugate is 225 In the case of Ac-DOTA-h11B6, the pharmaceutical composition may not contain any sucrose. According to certain embodiments, the composition comprises 225 Inclusion of sucrose in the Ac composition results in the formation of radiolytic degradation products, as described herein. Thus, in certain embodiments, sucrose may be excluded or limited to small amounts, e.g., less than 1%, less than 0.5%, less than 0.1%, less than 0.05%, or less than 0.01%.
According to certain embodiments, the pharmaceutical composition is free of any dextrins (e.g., cyclodextrins), monosaccharides, disaccharides, oligosaccharides, or polysaccharides.
According to certain embodiments, the pharmaceutical composition does not contain any mono-or disaccharides.
According to certain embodiments, the pharmaceutical composition does not contain any disaccharides.
According to certain embodiments, the pharmaceutical composition does not contain any sugar alcohol (e.g., sorbitol).
According to certain embodiments, the pharmaceutical composition is free of any cryoprotectant (e.g., sugar alcohol, glycerol, ethylene glycol, propylene glycol, dimethyl sulfoxide, etc.).
In certain embodiments, the pharmaceutical composition contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate and about 0.5% w/v% sodium ascorbate. In other embodiments, the pharmaceutical composition contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate and about 0.04w/v% polysorbate 20. In other embodiments, the pharmaceutical composition contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate and about 25mM to 27mM acetate buffer. In other embodiments, the medicament contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate, about 0.5w/v% sodium ascorbate, and about 0.04w/v% polysorbate 20. In other embodiments, the drug contains about 0.5mg/mL of the radioconjugate and conjugate intermediate, about 0.5w/v% sodium ascorbate, and about 25mM to 27mM acetate buffer. In other embodiments, the drug contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate, about 0.04w/v% polysorbate 20, and about 25mM to 27mM acetate buffer. In other embodiments, the pharmaceutical composition contains about 0.5mg/mL of the radioactive conjugate and conjugate intermediate, about 0.5w/v% sodium ascorbate, about 0.04w/v% polysorbate 20, and about 25mM to 27mM acetate buffer.
In certain aspects, the pharmaceutical composition contains a radioactive conjugate and an acetate buffer. In other aspects, the pharmaceutical composition contains a radioactive conjugate, an acetate buffer, and sodium ascorbate. In other aspects, the pharmaceutical composition contains a radioactive conjugate and polysorbate 20. In other aspects, the pharmaceutical composition contains a radioactive conjugate, sodium ascorbate, polysorbate 20, and an acetate buffer. In other aspects, the pharmaceutical composition comprises 225 Ac-DOTA-h11B6, acetate buffer. In other aspects, the pharmaceutical composition comprises 225 Ac-DOTA-h11B6, acetate buffer and sodium ascorbate. In other aspects, the pharmaceutical composition comprises 225 Ac-DOTA-h11B6, polysorbate 20. In other aspects, the pharmaceutical composition comprises 225 Ac-DOTA-h11B6, sodium ascorbate, polysorbate 20 and acetate buffer. In other aspects, the pharmaceutical composition comprises 225 Ac-TOPA-h11B6, acetate buffer. In other aspects, the pharmaceutical composition comprises 225 Ac-TOPA-h11B6, acetate buffer and sodium ascorbate. At other sideA composition comprising 225 Ac-TOPA-h11B6, polysorbate 20. In other aspects, the pharmaceutical composition comprises 225 Ac-TOPA-h11B6, sodium ascorbate, polysorbate 20 and acetate buffer.
According to one embodiment, the pharmaceutical composition contains a total amount of about 0.5mg/mL 225 A sterile aqueous solution of Ac-DOTA-h11B6 and DOTA-h11B6 in 25mM to 27mM (e.g., 25mM or 26.75 mM) acetate, 0.5% sodium ascorbate, and 0.04% polysorbate 20, preferably at a pH of about 5.5. In accordance with certain embodiments of the present invention, 225 radioactivity of Ac-DOTA-h11B6 225 The Ac dose was targeted to about 50 μci, about 100 μci, about 150 μci, or about 200 μci in 4mL (about 2mg of h11b6 mass) at the expected dosing time. In accordance with a further embodiment of the present invention, 225 radioactivity of Ac-DOTA-h11B6 225 The Ac dose is targeted to greater than 200 μci (e.g., about 250 μci or about 300 μci or about 350 μci, etc.) at the time of administration; for example, at the intended time of administration, the dose may comprise about 250 μCi in about 8mL or about 300 μCi in about 8mL or about 350 μCi in about 8mL (e.g., about 2mg of H11B6 or about 4mg or about 6mg or about 8mg or about 10mg mass per dose).
According to one embodiment, the pharmaceutical composition contains a total amount of about 0.5mg/mL 225 A sterile aqueous solution of Ac-TOPA-h11B6 and TOPA-h11B6 in 25mM to 27mM (e.g., 25mM or 26.75 mM) acetate, 0.5% sodium ascorbate, and 0.04% polysorbate 20, preferably at a pH of about 5.5. In accordance with certain embodiments of the present invention, 225 Radioactivity of Ac-TOPA-h11B6 225 The Ac dose was targeted to about 50 μci, about 100 μci, about 150 μci, or about 200 μci in 4mL (about 2mg of h11b6 mass) at the expected dosing time. In accordance with a further embodiment of the present invention, 225 radioactivity of Ac-TOPA-h11B6 225 The Ac dose is targeted to greater than 200 μci (e.g., about 250 μci or about 300 μci or about 350 μci, etc.) at the time of administration; for example, at the intended time of administration, the dose may comprise about 250 μCi in about 8mL or about 300 μCi in about 8mL or about 350 μCi in about 8mL (e.g., about 2mg of H11B6 or about 4mg or about 6mg or about 8mg or about 10mg mass per dose).
Implementation of enumeration
The following provides numbered exemplary embodiments of the present invention.
1. A method of treating cancer in a patient, the method comprising:
administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein:
the radioconjugate comprising at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2,
the radiometal complex comprises a radiometal and
upon administration, the radiometal provides targeted radioactivity of about 50 μCi to about 350 μCi per dose of the pharmaceutical composition.
A method of treating cancer in a patient, the method comprising:
administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein:
the radioconjugate comprising at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2,
the radiometal complex comprises a radiometal and
upon administration, the radiometal provides targeted radioactivity of about 350 μci to about 500 μci per dose of the pharmaceutical composition.
2. The method according to embodiment 1 or 1A, wherein the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2.
3. The method according to embodiment 2, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequences of SEQ ID NO. 1 and SEQ ID NO. 2 and SEQ ID NO. 3 and a light chain variable region; the light chain variable region comprises the amino acid sequences of SEQ ID NO. 4 and SEQ ID NO. 5 and SEQ ID NO. 6.
4. The method of embodiment 2 or 3, wherein the antibody comprises a heavy chain variable region (VH) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 9.
5. The method according to embodiment 2 or 3, wherein the antibody comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID No. 9.
6. The method according to any one of embodiments 2 to 5, wherein the antibody comprises a heavy chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 10 and a light chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 11.
7. The method according to any one of embodiments 2 to 5, wherein the antibody comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID NO. 10 and a light chain constant region comprising the amino acid sequence of SEQ ID NO. 11.
8. The method according to any one of embodiments 2 to 7, wherein the antibody comprises a heavy chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 12 and a light chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 13.
9. The method according to any one of embodiments 2 to 7, wherein the antibody comprises a heavy chain having the amino acid sequence of SEQ ID No. 12 and a light chain having the amino acid sequence of SEQ ID No. 13.
10. The method according to any one of embodiments 1 to 9 or 1A, wherein the radioactive metal is selected from the group consisting of: 225 Ac、 111 In、 177 Lu、 32 P、 47 Sc、 67 Cu、 77 As、 89 Sr、 90 Y、 99 Tc、 105 Rh、 109 Pd、 111 Ag、 131 I、 134 Ce、 149 Tb、 152 Tb、 155 Tb、 153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 255 fm sum 227 Th。
11. The method according to any one of embodiments 1 to 9 or 1A, wherein the radioactive metal is 225 Ac。
12. The method according to any one of embodiments 1 to 11 or 1A, wherein the radiometal complex comprises a chelator selected from the group consisting of: 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA), S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA), 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA), 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA), 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A), and derivatives thereof.
13. The method according to any one of embodiments 1 to 11 or 1A, wherein the radiometal complex comprises a chelator that is DOTA.
The method according to any one of embodiments 1 to 11 or 1A, wherein the radiometal complex comprises a chelator that is TOPA.
14. The method according to any one of embodiments 1 to 13 or 1A, wherein the radiometal complex comprises a chelate to DOTA 225 Ac。
The method according to any one of embodiments 1 to 11 or 1A, wherein the radiometal complex comprises a chelate to TOPA 225 Ac。
15. The method according to any one of embodiments 1 to 11 or 1A, wherein the radioconjugate comprises a radiometal chelated to: (a) A compound of formula (IV)
Or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and
R 4 is an antibody; or alternatively
(b) A compound of formula (V)
Or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and
R 4 is an antibody;
for example, wherein the chelator used to form the radioconjugate is a compound of the formula:
/>
16. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radioactive metal provides perAbout 2mg total antibody from about 25 μCi to about 350 μCi, or from about 50 μCi to about 350 μCi per about 2mg total antibody.
The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 350 μci per about 2mg total antibody to about 10mg total antibody (e.g., per about 4mg total antibody); or a targeted specific activity of about 50 to about 350 μci per about 2 to about 10mg total antibody (e.g., per about 4mg total antibody).
17. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 300 μci per about 2mg of total antibody, or about 50 μci to about 300 μci per about 2mg of total antibody.
The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 to about 300 μci per about 2 to about 10mg total antibodies (e.g., per about 4mg total antibodies); or a targeted specific activity of about 50 to about 300 μci per about 2 to about 10mg total antibody (e.g., per about 4mg total antibody).
18. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 250 μci per about 2mg of total antibody, or about 50 μci to about 250 μci per about 2mg of total antibody.
The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 250 μci per about 2mg total antibody to about 10mg total antibody (e.g., per about 4mg total antibody); or a targeted specific activity of about 50 to about 250 μci per about 2 to about 10mg total antibody (e.g., per about 4mg total antibody).
19. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radioactivityThe metal provides a targeted specific activity of about 25 μCi to about 200 μCi per about 2mg of total antibody, or about 50 μCi to about 200 μCi per about 2mg of total antibody.
20. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 150 μci per about 2mg of total antibody, or about 50 μci to about 150 μci per about 2mg of total antibody.
21. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 25 μci to about 100 μci per about 2mg of total antibody, or about 50 μci to about 100 μci per about 2mg of total antibody.
22. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 150 μci to about 250 μci per about 2mg total antibody.
23. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 50 μci per about 2mg total antibody.
24. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 100 μci per about 2mg total antibody.
25. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 150 μci per about 2mg total antibody.
26. The method according to any one of embodiments 2 to 15 or 13A or 14A, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 200 μci per about 2mg total antibody.
27. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises a targeted radioactivity concentration of about 1 μci/mL to about 100 μci/mL, or about 5 μci/mL to about 75 μci/mL, or about 10 μci/mL to about 60 μci/mL, or about 12.5 μci/mL to about 50 μci/mL, or about 12.5 μci/mL, or about 25 μci/mL, or about 37.5 μci/mL, or about 50 μci/mL.
28. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 1mg to about 5mg of total antibody, or about 1mg to about 4mg of total antibody.
The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 1mg to about 10mg total antibody, or about 2mg to about 8mg total antibody.
29. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 1mg to about 4mg of total antibody.
30. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 1mg to about 3mg of total antibody.
31. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 1.5mg to about 2.5mg total antibody.
32. The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 2mg total antibody.
The method according to any one of embodiments 2 to 26 or 13A or 14A or 16A or 17A or 18A, wherein the pharmaceutical composition comprises about 4mg total antibodies or about 8mg total antibodies.
33. The method of any one of embodiments 1 to 31 or 1A or 13A or 14A or 16A or 17A or 18A or 28A, wherein the one or more pharmaceutically acceptable excipients comprise one or more radioprotectants.
34. The method of embodiment 32 or 32A, wherein the one or more radioprotectants comprises sodium ascorbate, gentisic acid or a combination thereof.
35. The method of embodiment 32 or 32A, wherein the one or more radioprotectants comprises sodium ascorbate.
36. The method of embodiment 32 or 32A, wherein the one or more radioprotectants comprise gentisic acid.
37. The method of any one of embodiments 1 to 35 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the one or more pharmaceutically acceptable excipients further comprise one or more surfactants.
38. The method of embodiment 36, wherein the one or more surfactants comprise polysorbate 20.
39. The method of any one of embodiments 1 to 37 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the one or more pharmaceutically acceptable excipients further comprises an acetate buffer.
40. The method according to any one of embodiments 1 to 38 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition comprises a radioactive conjugate, sodium ascorbate, polysorbate 20, acetate buffer, and water (and optionally acetic acid for pH adjustment).
41. The method according to any one of embodiments 1 to 38 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition comprises an aqueous solution of a radioconjugate, about 24mM to 28mM acetate, about 0.25% to 0.75% sodium ascorbate, and about 0.01% to 0.15% polysorbate 20.
42. The method according to any one of embodiments 1 to 38 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition comprises an aqueous solution of a radioactive conjugate, about 25mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
43. The method according to any one of embodiments 1 to 38 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition comprises an aqueous solution of a radioactive conjugate, about 26.75mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
44. The method according to any one of embodiments 1 to 42 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition has a pH of about 5 to about 6 (e.g., about 5.5).
45. The method according to any one of embodiments 1 to 43 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition does not contain any preservative.
46. The method according to any one of embodiments 1 to 44 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition does not contain any sucrose.
47. The method according to any one of embodiments 1 to 44 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition does not contain any mono-, di-, oligo-or polysaccharides.
48. The method according to any one of embodiments 1 to 44 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition does not contain any mono-or disaccharides.
49. The method according to any one of embodiments 1 to 44 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition does not contain any disaccharides.
50. The method according to any one of embodiments 1 to 48 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition is stable at a temperature range of about 2 ℃ to 8 ℃ for at least about 72 hours, or at least about 96 hours, or at least about 120 hours.
51. The method according to any one of embodiments 2 to 49 or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the dose of the pharmaceutical composition has a volume of about 1mL to about 20mL, or about 1mL to about 10mL, or about 2mL to about 6mL, or about 3mL to about 5mL, or about 4 mL.
52. The method according to any one of embodiments 2 to 49 or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the dose of the pharmaceutical composition comprises about 2mg total antibody per about 4mL dose.
53. The method according to any one of embodiments 2 to 51 or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition comprises total antibodies in an amount of about 0.1mg/mL to 1.0mg/mL, or about 0.4mg/mL to 0.6mg/mL, or about 0.5 mg/mL.
54. The method according to any one of embodiments 2 to 52 or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the pharmaceutical composition further comprises a non-radiolabeled antibody (e.g., a conjugate intermediate such as DOTA-mAb, e.g., DOTA-h11B 6), wherein the non-radiolabeled antibody is the same antibody as the antibody conjugated to the radiometal complex.
55. The method of embodiment 53, wherein the total amount of the conjugated antibody and the non-radiolabeled antibody is not more than about 10mg, or about 9mg, or about 8mg, or about 7mg, or about 6mg, or about 5mg, or about 4mg, or about 3mg, or about 2mg.
56. The method according to any one of embodiments 1 to 54 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising intravenously administering the pharmaceutical composition to the patient.
57. The method according to any one of embodiments 1 to 55 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the pharmaceutical composition to the patient within about 168 hours, or within about 144 hours, or within about 120 hours, or within about 96 hours, or within about 72 hours, or within about 48 hours, or within about 24 hours, after chelating the radiometal to a conjugate intermediate to form the radioconjugate.
58. The method according to any one of embodiments 1 to 56 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the pharmaceutical composition once to the patient every about 4 weeks.
59. The method according to any one of embodiments 1 to 56 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the pharmaceutical composition to the patient once every about 8 weeks.
60. The method according to any one of embodiments 1 to 56 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the pharmaceutical composition to the patient once every about 12 weeks.
61. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is prostate cancer.
62. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is non-localized prostate cancer.
63. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is metastatic prostate cancer.
64. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is castration-resistant prostate cancer (CRPC).
65. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is metastatic castration-resistant prostate cancer (mCRPC).
66. The method according to any one of embodiments 1 to 59 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the cancer is mCRPC with adenocarcinoma.
67. The method according to any one of embodiments 1 to 65 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the patient has a testosterone castration level of about 50ng/dL or less.
68. The method of any one of embodiments 1 to 66 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the patient has been previously exposed to at least one Androgen Receptor (AR) targeted therapy.
69. The method of embodiment 67, wherein the AR-targeted therapy is abiraterone acetate, enzalutamide, apatamide, darunamine, or a combination of any of the foregoing.
70. The method according to any one of embodiments 1 to 68 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the patient has been previously subjected to chemotherapy.
71. The method of embodiment 69, wherein the chemotherapy involves administration of a taxane.
72. The method of any one of embodiments 1 to 70 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the patient has previously undergone orchiectomy or medical castration.
73. The method according to any one of embodiments 1 to 71 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, wherein the patient is receiving androgen deprivation therapy with a gonadotropin releasing hormone (GnRH) agonist or antagonist.
74. The method according to any one of embodiments 1 to 72 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the dose to the patient in a single administration.
75. The method of any one of embodiments 1 to 72 or 1A or 13A or 14A or 16A or 17A or 18A or 28A or 32A, comprising administering the dose in multiple administrations of more than one sub-dose.
75A. The method of embodiment 75, comprising administering the dose in two sub-doses (e.g., two 4mL sub-doses).
76. A pharmaceutical composition comprising:
a radio conjugate and one or more pharmaceutically acceptable excipients, wherein:
the radioconjugate comprises at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2, and
the radiometal complex comprises a radiometal.
77. The pharmaceutical composition of embodiment 76, wherein the one or more pharmaceutically acceptable excipients comprise one or more radioprotective agents.
78. The pharmaceutical composition according to embodiment 76 or 77, wherein the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2.
79. The pharmaceutical composition according to embodiment 78, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequences of SEQ ID No. 1 and SEQ ID No. 2 and SEQ ID No. 3; the light chain variable region comprises the amino acid sequences of SEQ ID NO. 4 and SEQ ID NO. 5 and SEQ ID NO. 6.
80. The pharmaceutical composition of embodiment 78 or embodiment 79, wherein the antibody comprises a heavy chain variable region (VH) having at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) having at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 9.
81. The pharmaceutical composition according to embodiment 78 or embodiment 79, wherein the antibody comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID No. 9.
82. The pharmaceutical composition according to any one of embodiments 78 to 81, wherein the antibody comprises a heavy chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 10 and a light chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 11.
83. The pharmaceutical composition according to any one of embodiments 78 to 81, wherein the antibody comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID No. 10 and a light chain constant region comprising the amino acid sequence of SEQ ID No. 11.
84. The pharmaceutical composition according to any one of embodiments 78 to 83, wherein the antibody comprises a heavy chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 12 and a light chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 13.
85. The pharmaceutical composition according to any one of embodiments 78 to 83, wherein the antibody comprises a heavy chain having the amino acid sequence of SEQ ID No. 12 and a light chain having the amino acid sequence of SEQ ID No. 13.
86. The pharmaceutical composition according to any one of embodiments 76-85, wherein the radioactive metal is selected from the group consisting of: 225 Ac、 111 In、 177 Lu、 32 P、 47 Sc、 67 Cu、 77 As、 89 Sr、 90 Y、 99 Tc、 105 Rh、 109 Pd、 111 Ag、 131 I、 134 Ce、 149 Tb、 152 Tb、 155 Tb、 153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 255 fm sum 227 Th。
87. The pharmaceutical composition of any one of embodiments 76-85, wherein the radiometal is 225 Ac。
88. The pharmaceutical composition according to any one of embodiments 76-87, wherein the radiometal complex comprises a chelator selected from the group consisting of: 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA), S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA), 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA), 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA), 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A), and derivatives thereof.
89. The pharmaceutical composition according to any one of embodiments 76-87, wherein the radiometal complex comprises a chelator that is DOTA.
90. The pharmaceutical composition of any one of embodiments 76-89, wherein the radiometal complex comprises a chelate to DOTA 225 Ac。
91. The pharmaceutical composition according to any one of embodiments 76-87, wherein the radioconjugate comprises a radiometal chelated to: (a) A compound of formula (IV)
Or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and
R 4 is an antibody; or alternatively
(b) A compound of formula (V)
Or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and
R 4 is an antibody;
for example, wherein the chelator used to form the radioconjugate is a compound of the formula:
92. the pharmaceutical composition of any one of embodiments 77-91, wherein the one or more radioprotectants comprises sodium ascorbate, gentisic acid, or a combination thereof (e.g., in an amount of about 0.1w/v% to about 5w/v%, or about 0.1w/v% to about 4w/v%, or about 0.1w/v% to about 3w/v%, about 0.1w/v% to about 2w/v%, or about 0.1w/v% to about 1w/v%, or about 0.25w/v% to about 0.75w/v%, or about 0.5 w/v%).
93. The pharmaceutical composition of any one of embodiments 77-91, wherein the one or more radioprotectants comprises sodium ascorbate (e.g., in an amount of about 0.1w/v% to about 5w/v%, or about 0.1w/v% to about 4w/v%, or about 0.1w/v% to about 3w/v%, about 0.1w/v% to about 2w/v%, or about 0.1w/v% to about 1w/v%, or about 0.25w/v% to about 0.75w/v%, or about 0.5 w/v%).
94. The pharmaceutical composition of any one of embodiments 77-91, wherein the one or more radioprotectants comprises gentisic acid (e.g., in an amount of about 0.1w/v% to about 5w/v%, or about 0.1w/v% to about 4w/v%, or about 0.1w/v% to about 3w/v%, about 0.1w/v% to about 2w/v%, or about 0.1w/v% to about 1w/v%, or about 0.25w/v% to about 0.75w/v%, or about 0.5 w/v%).
95. The pharmaceutical composition according to any one of embodiments 76-94, wherein the one or more pharmaceutically acceptable excipients further comprise one or more surfactants.
96. The pharmaceutical composition of embodiment 95, wherein the one or more surfactants comprise polysorbate 20.
97. The pharmaceutical composition according to any one of embodiments 76-96, wherein the one or more pharmaceutically acceptable excipients further comprises an acetate buffer.
98. The pharmaceutical composition of any one of embodiments 76-97, comprising the radioconjugate, sodium ascorbate, polysorbate 20, acetate buffer, and water (and optionally acetic acid to adjust pH).
99. The pharmaceutical composition of any one of embodiments 76-97, comprising an aqueous solution of the radioconjugate, about 24mM to 28mM acetate, about 0.25w/v% to 0.75w/v% sodium ascorbate, and about 0.01w/v% to 0.15w/v% polysorbate 20.
100. The pharmaceutical composition of any one of embodiments 76-97, comprising an aqueous solution of the radioconjugate, about 25mM acetate, about 0.5w/v% sodium ascorbate, and about 0.04w/v% polysorbate 20.
101. The pharmaceutical composition of any one of embodiments 76-97, comprising an aqueous solution of the radioconjugate, about 26.75mM acetate, about 0.5w/v% sodium ascorbate, and about 0.04w/v% polysorbate 20.
102. The pharmaceutical composition according to any one of embodiments 76-101, wherein the pharmaceutical composition has a pH of about 5 to about 6 (e.g., about 5.5).
103. The pharmaceutical composition according to any one of embodiments 76 to 101, wherein the pharmaceutical composition does not contain any preservative.
104. The pharmaceutical composition according to any one of embodiments 76 to 103, wherein the pharmaceutical composition does not contain any sucrose.
105. The pharmaceutical composition according to any one of embodiments 76 to 103, wherein the pharmaceutical composition does not comprise any monosaccharides, disaccharides, oligosaccharides or polysaccharides.
106. The pharmaceutical composition according to any one of embodiments 76 to 103, wherein the pharmaceutical composition does not contain any mono-or disaccharides.
107. The pharmaceutical composition according to any one of embodiments 76 to 103, wherein the pharmaceutical composition does not contain any disaccharides.
108. The pharmaceutical composition according to any one of embodiments 76-103, wherein the one or more pharmaceutically acceptable excipients consist of or consist essentially of an acetate buffer, sodium ascorbate, and an aqueous solution of polysorbate 20.
109. The pharmaceutical composition according to any one of embodiments 76-108, wherein the pharmaceutical composition is formulated for intravenous administration.
110. The pharmaceutical composition according to any one of embodiments 76-109, wherein the pharmaceutical composition is stable at a temperature range of about 2 ℃ to 8 ℃ for at least 72 hours, or at least 96 hours, or at least 120 hours.
111. The pharmaceutical composition of any one of embodiments 77-110, wherein the radioactive conjugate comprises an average of about 1 to about 4, or about 2 to about 3, chelator molecules conjugated to the antibody.
112. According to the embodimentThe pharmaceutical composition of any one of cases 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 350 μci per about 2mg total antibody.
112A. The pharmaceutical composition according to any one of embodiments 77 to 111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 350 μci per about 2mg to about 10mg total antibody.
112b. the pharmaceutical composition according to any one of embodiments 77 to 111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 350 μci to about 500 μci per about 2mg to about 10mg total antibody.
113. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 300 μci per about 2mg total antibody.
113A the pharmaceutical composition according to any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 300 μci per about 2mg to about 10mg total antibody.
114. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 250 μci per about 2mg total antibody.
114A the pharmaceutical composition according to any one of embodiments 77 to 111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 250 μci per about 2mg to about 10mg total antibody.
115. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 200 μci per about 2mg total antibody.
116. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides every about 2mg of total antibodySpecific activity of about 50 μCi to about 150 μCi.
117. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 100 μci per about 2mg total antibody.
118. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 50 to about 200 μci per about 2mg total antibody when administered.
119. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 50 μci per about 2mg total antibody when administered.
120. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 100 μci per about 2mg total antibody when administered.
121. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 150 μci per about 2mg total antibody when administered.
122. The pharmaceutical composition of any one of embodiments 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 200 μci per about 2mg total antibody when administered.
122A. The pharmaceutical composition according to any of embodiments 77 to 111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 300 μci per about 4mg total antibody when administered.
123. The pharmaceutical composition of any one of embodiments 77-122, comprising about 1mg to about 20mg of total antibody.
124. The pharmaceutical composition of any one of embodiments 77-122, comprising about 1mg to about 10mg of total antibody.
125. The pharmaceutical composition of any one of embodiments 77-122, comprising about 1mg to about 5mg of total antibody.
126. The pharmaceutical composition of any one of embodiments 77-122, comprising about 2mg total antibody.
127. The pharmaceutical composition of any one of embodiments 77-122, comprising about 10mg of total antibody.
128. The pharmaceutical composition of any one of embodiments 77-127, comprising a total amount of about 0.1mg/mL to 1.0mg/mL of conjugate intermediate and the radioactive conjugate.
129. The pharmaceutical composition of any one of embodiments 77-127, comprising a total amount of about 0.4mg/mL to 0.6mg/mL of conjugate intermediate and the radioactive conjugate.
130. The pharmaceutical composition of any one of embodiments 77-127, comprising a total amount of about 0.5mg/mL of conjugate intermediate and the radioactive conjugate.
131. The pharmaceutical composition of any one of embodiments 77-127, further comprising a non-radiolabeled antibody (e.g., a conjugate intermediate, such as DOTA-mAb, e.g., DOTA-h11B 6), wherein the non-radiolabeled antibody is the same antibody as the antibody conjugated to the radiometal complex.
132. The pharmaceutical composition of embodiment 131, wherein the total amount of the conjugated antibody and the non-radiolabeled antibody is no more than about 10mg, or about 9mg, or about 8mg, or about 7mg, or about 6mg, or about 5mg, or about 4mg, or about 3mg, or about 2mg.
133. A method for treating cancer in a patient, the method comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of any one of embodiments 76 to 132, or 112A, or 112B, or 113A, or 114A, or 122A.
134. The method according to embodiment 133, comprising administering the pharmaceutical composition once to the patient every about 4 weeks.
135. The method according to embodiment 133, comprising administering the pharmaceutical composition once to the patient every about 8 weeks.
136. The method according to embodiment 133, comprising administering the pharmaceutical composition once to the patient every about 12 weeks.
137. The method according to any one of embodiments 133-136, wherein the cancer is prostate cancer.
138. The method according to any of embodiments 133-136, wherein the cancer is non-localized prostate cancer.
139. The method according to any one of embodiments 133-136, wherein the cancer is metastatic prostate cancer.
140. The method according to any one of embodiments 133-136, wherein the cancer is castration-resistant prostate cancer (CRPC).
141. The method according to any one of embodiments 133-136, wherein the cancer is metastatic castration-resistant prostate cancer (mCRPC).
142. The method according to any of embodiments 133-136, wherein the cancer is mCRPC with an adenocarcinoma.
143. The method according to any of embodiments 133-142, wherein the patient has a testosterone castration level of about 50ng/dL or less.
144. The method according to any one of embodiments 133-143, wherein the patient has been previously exposed to at least one Androgen Receptor (AR) targeted therapy.
145. The method of embodiment 144, wherein the AR-targeted therapy is abiraterone acetate, enzalutamide, apatamide, darunamine, or a combination of any of the foregoing.
146. The method according to any of embodiments 133-145, wherein the patient has previously undergone chemotherapy.
147. The method of embodiment 146, wherein the chemotherapy involves administration of a taxane.
148. The method according to any of embodiments 133-147, wherein the patient has previously undergone orchiectomy or medical castration.
149. The method according to any of embodiments 133-148, wherein the patient is receiving androgen ablation therapy with a gonadotropin releasing hormone (GnRH) agonist or antagonist.
150. The pharmaceutical composition according to any one of embodiments 76 to 132 or 112A or 112B or 113A or 114A or 122A for use in the treatment of cancer; for example, prostate cancer, such as mCRPC.
151. A method of preparing the pharmaceutical composition according to any one of embodiments 76 to 132 or 112A or 112B or 113A or 114A or 122A, the method comprising combining a first intermediate composition and a second intermediate composition to form the pharmaceutical composition, wherein: the first intermediate composition comprises a radioactive conjugate and the second intermediate composition comprises a conjugate intermediate and does not comprise any radioactive conjugate.
152. The method of embodiment 151, wherein the radioactive conjugate and the conjugate intermediate comprise the same antibody.
153. The method of embodiment 151, wherein the radioactive conjugate and the conjugate intermediate comprise the same antibody and the same chelator.
154. The method according to any one of embodiments 151-153, wherein the first intermediate composition and the second intermediate composition comprise the same pharmaceutically acceptable excipient.
155. The method according to any one of embodiments 151-153, wherein the first intermediate composition and the second intermediate composition comprise the same pharmaceutically acceptable excipient in the same amount or substantially the same amount.
156. The method according to any one of embodiments 151-155, further comprising chelating the radiometal to a conjugate intermediate to form the radioconjugate.
According to a particular embodiment, including any one of embodiments 1 to 156 or 16A, 17A, 18A, 28A, 32A, 75A, 112B, 113A, 114A or 122A enumerated above, the radioactive conjugate is 225 Ac-DOTA-h11B6。
According to particular embodiments, embodiments 1 to 156 or 16A, 17A, 18 including the enumeration above A. 28A, 32A, 75A, 112B, 113A, 114A or 122A, the radioactive conjugate being TOPA- [ C7]Phenylthiourea-h 11B6 antibody conjugates, such as 225 Ac-TOPA-h11B6 (e.g., as shown in FIGS. 6A-6C).
The following examples are intended to further illustrate the nature of the invention. It should be understood that the following examples do not limit the present invention.
Examples
The h11B6 antibodies used in the examples below comprise a heavy chain according to SEQ ID NO. 12 and a light chain according to SEQ ID NO. 13.
111 Example 1: phase 0 imaging study of In-DOTA-h11B6 In humans
Proceeding with 111 A first human phase 0 imaging study of In-DOTA-h11B6 to determine the radioimmunotherapeutic potential of targeting hK2 In subjects with advanced prostate cancer. (clinical trial identifier NCT 04116164).
2Mg[111In]DOTA-h11B6 (nominal 185MBq [111 In)]) Is administered intravenously with or without 8mg of h11b 6. The formulation administered to the patient was 0.5mg/mL 111 25mM acetate of In-DOTA-h11B6, 8.5% sucrose (w/v), 0.04% polysorbate 20 (w/v), pH 5.5. Ultraviolet and radioactive HPLC chromatograms of the formulations were obtained. See fig. 1A and 1B.
Patients were observed for Adverse Events (AEs) for at least 2 weeks. Continuous gamma camera imaging including at least one SPECT/CT scan is performed until 8 days after administration. Serial blood samples were obtained over 2 weeks to determine serum radioactivity and h11B6 protein levels. Dosimetry of normal organs was estimated using olinoda-EXM.
The results of the first 6 patients are summarized in table 1. Treatment was tolerated in all patients, without adverse events and without evidence of increased accumulation in any organ, including salivary glands. The initial distribution volume appears to be limited to the vascular compartment. Slow clearance of radioactivity from the vascular compartment was observed in all patients, progressively targeting skeletal and non-skeletal lesions. H11B6mAb localized to bone and soft tissue metastases did not have significant normal tissue uptake and retained salivary glands. Serum pharmacokinetics and critical normal organ (liver, spleen, kidney) biodistribution both showed substantially no difference in antibody biological behavior at antibody mass levels of 2mg and 10 mg.
Table 1: patient characteristics, administration amount and tumor targeting
Patient numbering PSA Tumor location mAb mass [111In](MBq) Targeting
1 19.73 Bone 2mg 218 Is that
2 22.58 Liver 2mg 221 Is that
3 39.33 Bone 2mg 202 Is that
4 4.96 Bone 10mg 206 Is that
5 49.39 Bone 10mg 196 Is that
6 N/A Node 10mg 193 Is that
225 Example 2: preparation of formulation "A" comprising Ac-DOTA-h11B6
To prepare a formulation comprising actinium conjugated to h11B6, the same formulation used in stage 0 was prepared, but with 225 Ac-DOTA-h11B6 substitution 111 In-DOTA-h11B6. Preparation "formulation A" was prepared containing 0.5mg/mL 225 26.75mM acetate of Ac-DOTA-h11B6, 8.5% sucrose (w/v) and 0.04% polysorbate 20 (w/v), pH 5.5. Radiolytic degradation products were observed in the uv and radioactive HPLC chromatograms of the formulation. See fig. 2A and 2B. Radiolytic decomposition degradation The product was identified as being produced by the radiolysis of sucrose in the formulation.
225 Example 3: preparation of formulation "B" comprising Ac-DOTA-h11B6
The cryoprotectant sucrose was eliminated from formulation a due to the formation of secondary radiolytic degradation products from the primary radiation. Changing form from a frozen solid to a liquid solution. However, elimination of sucrose results in 225 Accelerated degradation of Ac-DOTA-h11B6 pharmaceutical products, in particular of h11B6 antibodies. Sodium ascorbate (vitamin C) was added at 0.5% w/v as a sacrificial radioprotectant to attenuate degradation, resulting in a composition containing 0.5mg/mL 225 "formulation B" of Ac-DOTA-h11B6, 26.75mM acetate, 0.5% w/v sodium ascorbate (vitamin C) and 0.04% polysorbate 20, pH 5.5. Ultraviolet and radioactive HPLC chromatograms of the formulations were obtained. See fig. 3A to 3D. 225 Degradation of the Ac-DOTA-h11B6 drug product was significantly reduced in formulation B.
225 225 Example 4: preparation of Ac-DOTA-h11B6 and formulation "B" comprising Ac-DOTA-h11B6
Although this example describes a container comprising 225 Preparation of the pharmaceutical product Ac-DOTA-h11B6, but similar methods may also be used to prepare the pharmaceutical product containing 225 Ac-TOPA-h11B6 substitution 225 Pharmaceutical product of Ac-DOTA-h11B 6.
This example describes the preparation of 225 Ac-DOTA-h11B6 pharmaceutical products and solutions containing the same. Antibody h11B6 is described, for example, in us patent 10,100,125, which is incorporated herein by reference. Antibody h11B6 can also be prepared using the method described in U.S. patent 9,873,891, incorporated herein by reference, using a CHO DG44 derived cell line and hef1α promoter double gene vector, commercially available from Fujifilm Diosynth Biotechnologies.
After preculture and expansion, the cell culture can be clarified using known filtration techniques. The filtrate was concentrated and diafiltered in buffer (25mM NaOAc,pH 5.5) to a target final concentration of 10 g/L. H11B6 was filtered through a 0.2 μm filter, filled into sterile bags, and can be frozen at-65℃for long term storage prior to conjugation.
Thawed h11B6 was then diafiltered (buffer exchanged) to 50mM Bicine,120mM NaCl,pH 8.5 for subsequent conjugation of DOTA (1, 4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid) to h11B6. The retentate from the previous step was transferred to the reactor and stirred while warming to 25 ℃. An aqueous solution of p-SCN-Bn-DOTA was prepared and added to the reactor. The reaction was kept at 25℃for 20 hours. The product of the conjugation reaction (DOTA-h 11B 6) was transferred directly into the retentate vessel for final diafiltration with 25mM NaOAc,pH 5.5. Next, the DOTA-h11B6 conjugate intermediate was filtered through a 0.2 μm filter, filled into sterilized polycarbonate containers, and can be frozen at less than or equal to-65℃for long term storage.
The conjugation reaction resulted in the addition of multiple DOTA molecules to the epsilon amino groups of the lysine side chains of the h11B6 mAb. The ratio of conjugate to antibody (CAR), which represents the number of DOTA molecules per h11B6 mAb molecule, can be measured by complete mass analysis using RP-HPLC with online mass analysis. Based on the molecular structure of p-SCN-Bn-DOTA, each DOTA residue adds a 552Da mass to the antibody, which can be easily detected by complete mass analysis. To reduce sample complexity, DOTA-h11B6 conjugate intermediates were treated with PNGase F to remove N-linked glycans and carboxypeptidase B to remove C-terminal lysine residues. The average CAR of DOTA-h11B6 of 2.6 was calculated as a weighted average of all detected CAR classes.
225 Ac-DOTA-h11B6 pharmaceutical products are produced in a continuous operation from precursor DOTA-h11B6 conjugate intermediates which are conjugated to 225 Actinium trichloride reaction to give target specific Activity of 170. Mu. Ci/mg 225 Actinium-radiolabeled drug substance. Synthesis 225 The Ac-DOTA-h11B6 drug substance was purified by PD-10 column purification and formulated in situ. As described below, by 225 The Ac-DOTA-h11B6 drug substance was mixed with DOTA-h11B6 and reconstitution buffer to prepare four drug product forms (50. Mu. Ci, 100. Mu. Ci, 150. Mu. Ci and 200. Mu. Ci in 2mg protein). To be mixed with The products were separately sterile filtered and aseptically filled into final patient vials.
Intermediate purification buffers (26.75 mM acetate, 0.04% polysorbate 20, acetic acid, pH 5.5) were prepared for final multiplex purification buffer and can be stored at 2℃to 8℃.ltoreq.30 days before use. Sodium ascorbate was added to the intermediate purification buffer and filtered through a 0.2 μm sterile filter into a sterile product holding vessel to produce the final complex purification buffer (26.75 mM acetate, 0.5% (w/v) sodium ascorbate, 0.04% (w/v) polysorbate 20, acetic acid, pH 5.5).
The DOTA-h11B6 conjugate intermediate was thawed at room temperature. An actinium trichloride solution was prepared by dissolving actinium trinitrate in 0.1N hydrochloric acid (sources of Ac-225, already in the trichlorinated form, could also be used). Actinium trichloride (800. Mu. Ci to 1300. Mu. Ci) was incubated with 4.4mg DOTA-h11B6 and sodium acetate buffer (pH adjusted with acetic acid, prepared and stored in advance for. Ltoreq.6 months) and the pH adjusted to 6.5. Purification was then carried out on a pretreated PD-10 column 225 Ac-DOTA-h11B6 and eluted with final purification buffer. After purification, the amount of radioactivity was determined.
Four doses (50. Mu. Ci, 100. Mu. Ci, 150. Mu. Ci, and 200. Mu. Ci) were obtained in the preparation, and DOTA-h11B6 conjugate intermediate was reconstituted to 0.5mg/mL (26.75 mM acetate, 0.5% (w/v) sodium ascorbate, 0.04% (w/v) polysorbate 20, acetic acid, pH 5.5) using final reconstitution buffer and filtered through a 0.2 μm sterile filter. Then will 225 Ac-DOTA-h11B6 was dispensed into intermediate vials to achieve the desired unit dose (50. Mu. Ci, 100. Mu. Ci, 150. Mu. Ci, and 200. Mu. Ci in 4mL at the expected time of administration to the patient) and reconstituted DOTA-h11B6 conjugate intermediate was added to a volume of 6.8mL to produce the drug product. The drug product was then filtered through a 0.2 μm sterilizing filter and aseptically filled to a volume of 4.8 mL. The remaining drug product was also filtered through a 0.2 μm sterilizing filter and aseptically filled for release testing of the drug product. The pharmaceutical product is stored immediately at 2 ℃ to 8 ℃.
Thus, radiolabeled pharmaceutical products 225 Ac-DOTA-h11B6 was prepared as sterile solutionThe liquid is for intravenous injection and contains no preservative. At the desired time of administration to the patient, 225 Ac-DOTA-h11B6 was obtained in unit doses of four Drug Products (DP): 50. Mu. Ci, 100. Mu. Ci, 150. Mu. Ci and 200. Mu. Ci.
The targeted composition of the drug product is provided in table 2 below. The compositions may alternatively be used separately 225 Ac-TOPA-h11B6 and TOPA-h11B6 substitution 225 Ac-DOTA-h11B6 and DOTA-h11B 6.
Table 2: composition and concentration of 50 μCi, 100 μCi, 150 μCi and 200 μCi drug products
a target fill volume (4.8 mL) includes an additional 0.8mL.
b target activity concentration at correction.
c will 225 Ac-DOTA-h11B6 was combined with DOTA-h11B6 and reconstitution buffer to produce the respective unit doses of the pharmaceutical product as described herein.
Example 5A: DOTA-h11B6 stability study
This study was performed to monitor the stability profile of DOTA-h11B6 drug substance intermediate (10 mg/mL in 25mM acetate, pH adjusted to 5.5) under various environmental conditions and time periods. Study test articles were prepared by aliquoting Drug Substance Intermediate (DSI) into 20mL polycarbonate bottles at a fill volume of 9 mL.
Study parameters
Stability study results
Stability of DOTA-h11B6 DSI maintained under recommended, accelerated and two stress conditions results are shown below. All test parameter result values observed for each assay study at all time points of DSI maintained under recommended storage conditions exceeded criteria consistent with the most preferred embodiment of stability when maintained at a temperature of about-65 ℃ for about 4 years or more after storage for about 48 months or more, after storage for about 6 months or more at a temperature of about-40 ℃, after storage for about 6 months or more at a temperature of about 5 ℃. Of particular note, DSI maintained for 6 months under accelerated conditions (-40 ℃) and DSI maintained for 6 months under stressed conditions (5 ℃) showed results consistent with the preferred embodiment of stability maintained for about 4 years or more at-65 ℃.
The results of DOTA-h11B6 DSI maintained for 1 month under stress conditions (25 ℃) showed a lower degradation rate of drug substance intermediates exposed to the stress storage conditions.
-65℃data
Table a: stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-65 ℃.
Table a (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-65℃
/>
Table a (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-65℃
SE-HPLC = size exclusion high performance liquid chromatography; HMW = high molecular weight; LMW = low molecular weight; CE-SDS = sodium dodecyl sulfate capillary electrophoresis; lc=light chain; hc=heavy chain; nghc=non-glycosylated heavy chain; igG = immunoglobulin G; CIEF = cation exchange; nr=unreported
-40 ℃ data
Table B: stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-40℃
Table B (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-40℃
Table B (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at-40℃
SE-HPLC = size exclusion high performance liquid chromatography; HMW = high molecular weight; LMW = low molecular weight; CE-SDS = sodium dodecyl sulfate capillary electrophoresis; lc=light chain; hc=heavy chain; nghc=non-glycosylated heavy chain; igG = immunoglobulin G; cIEF = capillary isoelectric focusing; RP-HPLC = reverse phase high performance liquid chromatography
Data at 5 DEG C
Table C: stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 5℃
Table C (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 5℃
Table C (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 5℃
SE-HPLC = size exclusion high performance liquid chromatography; HMW = high molecular weight; LMW = low molecular weight; CE-SDS = sodium dodecyl sulfate capillary electrophoresis; lc=light chain; hc=heavy chain; nghc=non-glycosylated heavy chain; igG = immunoglobulin G; cIEF = capillary isoelectric focusing; RP-HPLC = reverse phase high performance liquid chromatography
25 ℃ data
Table D: stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 25℃
Table D (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 25℃
Table D (continuous): stability results of DOTA-h11B6 Drug Substance Intermediate (DSI) at 25℃
SE-HPLC = size exclusion high performance liquid chromatography; HMW = high molecular weight; LMW = low molecular weight; CE-SDS = sodium dodecyl sulfate capillary electrophoresis; lc=light chain; hc=heavy chain; nghc=non-glycosylated heavy chain; igG = immunoglobulin G; cIEF = capillary isoelectric focusing; RP-HPLC = reverse phase high performance liquid chromatography
111 Example 5B: in-DOTA-h11B6 stability study
The study was conducted to monitor 111 Stability properties of In-DOTA-h11B6 pharmaceutical product (DP) under recommended storage conditions. Research test articles were prepared by septum filling pharmaceutical products from pre-filled, capped and crimped 10R borosilicate vials.
Study parameters
Stability study results
111 Stability of In-DOTA-h11B6 DP maintained at recommended and accelerated conditions results are shown below. All test parameter result values observed for each assay study at all time points of DP maintained under recommended storage conditions exceeded criteria consistent with the most preferred embodiment of stability after storage at a temperature of about-40 ℃ for about 72 hours or more and/or after storage at a temperature of about 5 ℃ for about 72 hours or more.
DP maintained for 72 hours at acceleration (5 ℃) showed results consistent with the preferred embodiment of stability maintained for about 72 hours or more at-40 ℃.
40 ℃ data
Table A1: 111 stability results of In-DOTA-h116B6 on storage at-40 ℃
Table A1: (subsequent) 111 Stability results of In-DOTA-h116B6 on storage at-40 ℃
Nd=undetected, hmws=high molecular weight species, lmws=low molecular weight species
Data at 5 DEG C
Table A2: 111 stability results of In-DOTA-h116B6 on storage at 5℃
Table A2: (subsequent) 111 Stability results of In-DOTA-h116B6 on storage at 5℃
HMWS = high molecular weight species, LMWS = low molecular species, μci = microcurie; nd=undetected
225 Example 5C: ac-DOTA-h11B6 stability study
The study was conducted to monitor 225 Stability properties of Ac-DOTA-h11B6 drug products (50. Mu. Ci and 200. Mu. Ci) under recommended storage conditions. Research test articles were prepared by filling the drug product with the septum of a pre-sealed 10R cyclic olefin polymer vial.
Study parameters
Stability study results
225 In-DOTA-h11B6 DP was maintained at recommended storageStability under conditions results are shown below. All test parameter result values observed for each assay study at all time points of DP maintained under recommended storage conditions exceeded criteria consistent with the most preferred embodiment of stability when maintained after about 96 hours of storage.
Data at 2℃to 8 ℃
Table B1: 225 stability results of Ac-DOTA-h116B6 stored at 2℃to 8 ℃ (50. Mu. Ci)
Table B1: stability results of (follow) 225Ac-DOTA-h116B6 stored at 2℃to 8 ℃ (50. Mu. Ci)
/>
SEC = size exclusion chromatography, HMWS = high molecular weight species, LMWS = low molecular weight species, μci = microcurie;
Table B2: 225 stability results of Ac-DOTA-h116B6 stored at 2℃to 8 ℃ (200. Mu. Ci)
Table B2: stability results of (follow) 225Ac-DOTA-h116B6 stored at 2℃to 8 ℃ (200. Mu. Ci)
SEC = size exclusion chromatography, HMWS = high molecular weight species, LMWS = low molecular weight species, μci = microcurie;
example 6: use of actinium-225-labeled antibodies targeting human kallikrein-2 (hK 2) for advanced prostate cancer (accession number: NCT04644770;69086420PCR 1001)
This example describes a first human phase 1 study to evaluate 225 Safety, pharmacokinetics, pharmacodynamics and preliminary anti-tumor activity of Ac-DOTA-h11B6 administered to adult patients with mCRPC who have disease progression at or after AR targeted therapy. Formulation B described in examples 3 and 4 was administered to patients in phase 1 trial. As discussed herein in the context of the present disclosure, 225 Ac-DOTA-h11B6 is a hK 2-specific monoclonal antibody h11B6 that is labeled with DOTA and sequestered to an alpha-microparticle radionuclide 225 Ac, and is radioimmunotherapy targeting the hK2 antigen. It should be noted that a patient cohort for the present study will be administered according to a similar clinical method as described in this example and using a similar pharmaceutical composition 225 Ac-TOPA-h11B6 substitution 225 Ac-DOTA-h11B6。
The main purpose is to determine 225 Safety of Ac-DOTA-h11B6 and recommended phase 2 dose (RP 2D) as well as evaluation of incidence, duration and severity of adverse events, including Dose Limiting Toxicity (DLT). Secondary objectives and endpoints will evaluate primary anti-tumor activity and provide for a response to 225 The pharmacology of Ac-DOTA-h11B6 is further understood. See, e.g., table 3.
TABLE 3 Table 3
Will be 225 Ac-DOTA-h11B6 was administered to adult males older than 18 years with mCRPC, which had previously been exposed to at least one novel AR targeting therapy. 225 The administration of Ac-DOTA-h11B6 will be performed in 2 parts: dose escalation (part 1) and dose extension (part 2).
The response to treatment was assessed according to the PCWG3 response criteria.
Blood samples were collected to characterize the pharmacokinetics of serum radioactivity and the concentration of h11B6 antibodies, and characterized 225 The presence of an anti-drug antibody to Ac-DOTA-h11B 6.
225 The safety of Ac-DOTA-h11B6 will be assessed by physical examination, eastern Cooperative Oncology Group (ECOG) performance status, electrocardiogram, clinical laboratory tests, vital signs, and adverse event monitoring. An echocardiogram or multi-gated acquisition scan will be evaluated at screening; if indicated clinically, a subsequent evaluation will be made. The severity of adverse events will be assessed using the "national cancer institute adverse event common terminology standard" (version 5.0). Concomitant drug use will be noted.
Dose escalation decisions will be supported by an improved continuous re-assessment method (mcmr) based on Bayesian Logistic Regression Model (BLRM) and controlling overdose (EWOC).
Inclusion criteria included the following
Each potential patient must meet all of the following criteria:
the exclusion criteria included the following
Any potential patient meeting any of the following criteria will be excluded.
A. Part 1: dose escalation
The participants will accept 225 Intravenous (IV) injection of Ac-DOTA-h11B6, one or more doses, as described below.
In section 1, 50. Mu. Ci/2mg 225 Ac-DOTA-h11B6 was administered to the first dose ascending queue. Radioactivity after DLT evaluation in this initial queue 225 The dose escalation of Ac-DOTA-h11B6 to the next dose level will be based on a review of all available additional data, including but not limited toPharmacokinetics, pharmacodynamics, safety and primary antitumor activity.
Table 4 shows a planned (temporary) dose escalation schedule to illustrate possible dose escalation routes that include doses above 200 μci (e.g., 300 μci or higher). Intermediate dose level increments may ensure safety of study participants. The initial cohort will receive 50. Mu. Ci of radioactivity 225 Ac-DOTA-h11B6. The incrementing initially occurs in 50 μCi increments. A dosing interval of one dose per 8 weeks will be used. The mass of the starting antibody (h 11B 6) was 2mg, possibly increased to 10mg. Initially, the antibody mass dose will remain constant as the radioactivity between the cohorts increases.
Table 4: dose escalation protocol
In this study, the final drug product to be administered to the participants had two components: 225 Ac-DOTA-h11B6 and unlabeled DOTA-h11B6 antibodies. The two components may be pre-mixed in a single vial. Both components will be provided at the prescribed radioactive dose and a total antibody mass of between 2mg and 10mg at each participant visit. See table 5.
225 Table 5: ac-DOTA-h11B6 radiation therapy administration
225Ac-DOTA-h11B6 radiological study product was a single use, sterile, chilled solution for injection in a cycloolefin polymer vial closed with a latex-free stopper and an aluminum seal. 225Ac-DOTA-h11B6 was formulated in 26.75mM acetate, 0.5% sodium ascorbate and 0.04% polysorbate 20 in sterile aqueous solution (pH 5.5). The products studied were clear, colorless to pale yellow, and free of visible particulate matter. 225Ac-DOTA-h11B6 vials were stored refrigerated at a temperature ranging from 2℃to 8℃and kept away And (5) preserving the light. The pharmaceutical product does not contain any preservative and is designed for single use only. The vials supplied to the clinic contained an additional 0.8mL (4.8 mL total) in order to ultimately withdraw a dose of 4.0±0.4mL, depending on the actual administration time. The radioactivity concentration of 225Ac-DOTA-h11B6 was initially directed to 50 μci, 100 μci, 150 μci, or 200 μci in 4mL (2 mg), followed by doses higher than 200 μci (e.g., 300 μci, which would have a total volume of 8mL, with a protein concentration of 2mg/4mL, thus 4mg total protein at the expected time of administration). Intermediate dose level increments may ensure safety of study participants. As described above, for additional patient cohorts, a similar study product for this study will contain 225 Ac-TOPA-h11B6 substitution 225 Ac-DOTA-h11B6。
Dose escalation is supported using an adaptive dose escalation strategy guided by a modified continuous re-assessment approach based on BLRM and EWOC.
RP2D was determined after reviewing all available pharmacokinetic, pharmacodynamic, safety and efficacy data. Once RP2D is determined, the patient will be treated to confirm 225 Safety, pharmacokinetics, pharmacodynamics and preliminary antitumor activity of Ac-DOTA-h11B6 against RP2D in fraction 2.
B. Part 2: dose expansion
In section 2, the same as that measured in section 1 will be used 225 RP2D of Ac-DOTA-h11B6 is administered to patients in one or more cohorts.
All adverse events and adverse events meeting the DLT criteria will be reviewed and confirmed. Adverse events will be evaluated according to NCI CTCAE version 5.0. The criteria for DLT are summarized in table 6.
As described above, according to a similar clinical approach described in this example, a patient cohort for this study will be administered 225 Ac-TOPA-h11B6 substitution 225 Ac-DOTA-h11B6。
a Table 6: dose limiting toxicity criteria
/>
The result index is provided in table 7.
Table 7: results index
The secondary results indicators are provided in table 8.
Table 8: second-level result index
/>
Clinical outcome in stage
For 23 participants with metastatic castration-resistant prostate cancer (mCRPC), in a 69086420PCR1001 study, administration was performed at 4 radioactive dose levels of 50 μci, 100 μci, 150 μci, and 200 μci 225 The median number of doses received was 2 doses (range: 1 to 6) and the median treatment duration was 1.87 months (range: 1 to 10.8). Dose Limiting Toxicity (DLT) was not reported at any of the 4 radioactive dose levels.
For those participants, the most frequently reported (15% or more) treatment-emergent adverse events (TEAE) were fatigue (39.1%), loss of appetite (34.8%), diarrhea (26.1%), anemia and thrombocytopenia (21.7% each) and nausea and leukopenia (17.4% each). Most of these commonly reported TEAEs are grade 1 or grade 2, except that 1 participant is grade 3 fatigued at 50 μci,1 participant is grade 4 thrombocytopenia at 150 μci, and 2 participants (1 at 50 μci,1 at 200 μci) are grade 3 anaemia. Treatment-induced Serious Adverse Events (SAE) have been reported for 2 participants: 1 participant was hypokalemia at 100. Mu. Ci, and 1 participant was hypocalcemia at 150. Mu. Ci. One (1) participant was interrupted at 150 μCi due to thrombocytopenia, while all other interrupted participants were due to progressive disease or other causes. No dose reduction is required in any of the participants. No mortality in the treatment was observed. Efficacy signals in these participants include, for example, a 50% or more decrease in PSA from baseline in patients with a radiation dose greater than or equal to 100 uCi.
The disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated by reference in their entirety for all purposes.
Sequence listing
<110> Janssen Biotech Co., inc. (Janssen Biotech, inc.)
<120> compositions and methods for treating prostate cancer
<130> JBI6423WOPCT1
<150> US 63/193,704
<151> 2021, 5, 27 days
<150> US 63/335,761
<151> 2022, 4, 28 days
<160> 15
<170> version of Patentin3.5
<210> 1
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 1
Ser Asp Tyr Ala Trp Asn
1 5
<210> 2
<211> 16
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 2
Tyr Ile Ser Tyr Ser Gly Ser Thr Thr Tyr Asn Pro Ser Leu Lys Ser
1 5 10 15
<210> 3
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 3
Gly Tyr Tyr Tyr Gly Ser Gly Phe
1 5
<210> 4
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 4
Lys Ala Ser Glu Ser Val Glu Tyr Phe Gly Thr Ser Leu Met His
1 5 10 15
<210> 5
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 5
Ala Ala Ser Asn Arg Glu Ser
1 5
<210> 6
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 6
Gln Gln Thr Arg Lys Val Pro Tyr Thr
1 5
<210> 7
<211> 261
<212> PRT
<213> Homo sapiens (Homo sapiens)
<220>
<223> hK2 sequence
<400> 7
Met Trp Asp Leu Val Leu Ser Ile Ala Leu Ser Val Gly Cys Thr Gly
1 5 10 15
Ala Val Pro Leu Ile Gln Ser Arg Ile Val Gly Gly Trp Glu Cys Glu
20 25 30
Lys His Ser Gln Pro Trp Gln Val Ala Val Tyr Ser His Gly Trp Ala
35 40 45
His Cys Gly Gly Val Leu Val His Pro Gln Trp Val Leu Thr Ala Ala
50 55 60
His Cys Leu Lys Lys Asn Ser Gln Val Trp Leu Gly Arg His Asn Leu
65 70 75 80
Phe Glu Pro Glu Asp Thr Gly Gln Arg Val Pro Val Ser His Ser Phe
85 90 95
Pro His Pro Leu Tyr Asn Met Ser Leu Leu Lys His Gln Ser Leu Arg
100 105 110
Pro Asp Glu Asp Ser Ser His Asp Leu Met Leu Leu Arg Leu Ser Glu
115 120 125
Pro Ala Lys Ile Thr Asp Val Val Lys Val Leu Gly Leu Pro Thr Gln
130 135 140
Glu Pro Ala Leu Gly Thr Thr Cys Tyr Ala Ser Gly Trp Gly Ser Ile
145 150 155 160
Glu Pro Glu Glu Phe Leu Arg Pro Arg Ser Leu Gln Cys Val Ser Leu
165 170 175
His Leu Leu Ser Asn Asp Met Cys Ala Arg Ala Tyr Ser Glu Lys Val
180 185 190
Thr Glu Phe Met Leu Cys Ala Gly Leu Trp Thr Gly Gly Lys Asp Thr
195 200 205
Cys Gly Gly Asp Ser Gly Gly Pro Leu Val Cys Asn Gly Val Leu Gln
210 215 220
Gly Ile Thr Ser Trp Gly Pro Glu Pro Cys Ala Leu Pro Glu Lys Pro
225 230 235 240
Ala Val Tyr Thr Lys Val Val His Tyr Arg Lys Trp Ile Lys Asp Thr
245 250 255
Ile Ala Ala Asn Pro
260
<210> 8
<211> 117
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 8
Gln Val Gln Leu Gln Glu Ser Gly Pro Gly Leu Val Lys Pro Ser Asp
1 5 10 15
Thr Leu Ser Leu Thr Cys Ala Val Ser Gly Asn Ser Ile Thr Ser Asp
20 25 30
Tyr Ala Trp Asn Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp
35 40 45
Ile Gly Tyr Ile Ser Tyr Ser Gly Ser Thr Thr Tyr Asn Pro Ser Leu
50 55 60
Lys Ser Arg Val Thr Met Ser Arg Asp Thr Ser Lys Asn Gln Phe Ser
65 70 75 80
Leu Lys Leu Ser Ser Val Thr Ala Val Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Thr Gly Tyr Tyr Tyr Gly Ser Gly Phe Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser
115
<210> 9
<211> 111
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 9
Asp Ile Val Leu Thr Gln Ser Pro Asp Ser Leu Ala Val Ser Leu Gly
1 5 10 15
Glu Arg Ala Thr Ile Asn Cys Lys Ala Ser Glu Ser Val Glu Tyr Phe
20 25 30
Gly Thr Ser Leu Met His Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro
35 40 45
Lys Leu Leu Ile Tyr Ala Ala Ser Asn Arg Glu Ser Gly Val Pro Asp
50 55 60
Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser
65 70 75 80
Ser Leu Gln Ala Glu Asp Val Ala Val Tyr Tyr Cys Gln Gln Thr Arg
85 90 95
Lys Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 10
<211> 330
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 10
Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys
1 5 10 15
Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr
20 25 30
Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser
35 40 45
Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser
50 55 60
Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr
65 70 75 80
Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys
85 90 95
Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys
100 105 110
Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro
115 120 125
Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys
130 135 140
Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp
145 150 155 160
Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu
165 170 175
Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu
180 185 190
His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn
195 200 205
Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly
210 215 220
Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu
225 230 235 240
Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr
245 250 255
Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn
260 265 270
Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe
275 280 285
Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn
290 295 300
Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr
305 310 315 320
Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
325 330
<210> 11
<211> 107
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 11
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
1 5 10 15
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
20 25 30
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
35 40 45
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
50 55 60
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
65 70 75 80
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
85 90 95
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
100 105
<210> 12
<211> 447
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 12
Gln Val Gln Leu Gln Glu Ser Gly Pro Gly Leu Val Lys Pro Ser Asp
1 5 10 15
Thr Leu Ser Leu Thr Cys Ala Val Ser Gly Asn Ser Ile Thr Ser Asp
20 25 30
Tyr Ala Trp Asn Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp
35 40 45
Ile Gly Tyr Ile Ser Tyr Ser Gly Ser Thr Thr Tyr Asn Pro Ser Leu
50 55 60
Lys Ser Arg Val Thr Met Ser Arg Asp Thr Ser Lys Asn Gln Phe Ser
65 70 75 80
Leu Lys Leu Ser Ser Val Thr Ala Val Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Thr Gly Tyr Tyr Tyr Gly Ser Gly Phe Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu
115 120 125
Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys
130 135 140
Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser
145 150 155 160
Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser
165 170 175
Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser
180 185 190
Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn
195 200 205
Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His
210 215 220
Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val
225 230 235 240
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
245 250 255
Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu
260 265 270
Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
275 280 285
Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser
290 295 300
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
305 310 315 320
Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile
325 330 335
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
340 345 350
Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
355 360 365
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
370 375 380
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
385 390 395 400
Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg
405 410 415
Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
420 425 430
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
435 440 445
<210> 13
<211> 218
<212> PRT
<213> artificial sequence
<220>
<223> synthetic polypeptide
<400> 13
Asp Ile Val Leu Thr Gln Ser Pro Asp Ser Leu Ala Val Ser Leu Gly
1 5 10 15
Glu Arg Ala Thr Ile Asn Cys Lys Ala Ser Glu Ser Val Glu Tyr Phe
20 25 30
Gly Thr Ser Leu Met His Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro
35 40 45
Lys Leu Leu Ile Tyr Ala Ala Ser Asn Arg Glu Ser Gly Val Pro Asp
50 55 60
Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser
65 70 75 80
Ser Leu Gln Ala Glu Asp Val Ala Val Tyr Tyr Cys Gln Gln Thr Arg
85 90 95
Lys Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys Arg
100 105 110
Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln
115 120 125
Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr
130 135 140
Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser
145 150 155 160
Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr
165 170 175
Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys
180 185 190
His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro
195 200 205
Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 14
<211> 351
<212> DNA
<213> artificial sequence
<220>
<223> synthetic DNA
<400> 14
caggttcagc tgcaggaaag cggacctggc ttggtgaaac ccagcgatac ccttagcctg 60
acatgtgctg tgtctggcaa ttccatcact tccgactatg cgtggaactg gattcggcaa 120
ccaccgggaa aagggctcga gtggataggg tacatcagct attctggttc aaccacgtac 180
aatccctcac tgaagagtag ggttaccatg tccagagaca cctccaagaa ccagttcagc 240
ctgaagctga gtagtgtgac agccgtagat acagccgtct attactgcgc aacagggtac 300
tactatggct ctggcttttg gggtcaagga actctcgtca ctgtgtcaag c 351
<210> 15
<211> 333
<212> DNA
<213> artificial sequence
<220>
<223> synthetic DNA
<400> 15
gacatagtgc tcactcagag ccctgatagc ttggctgtca gtcttgggga aagagccacc 60
atcaactgca aagcgtccga aagcgtcgag tatttcggga ctagcctgat gcactggtat 120
cagcagaaac ccggacaacc gcctaagctg ctgatctatg cagcctctaa tcgcgaaagt 180
ggcgttccag acaggttttc cggttctgga tcaggcacag acttcaccct cacgatttcc 240
tcactgcaag ctgaggatgt agccgtgtac tactgtcagc agacacggaa agtgccctac 300
acctttggtc agggcacaaa gctggagatt aag 333

Claims (149)

1. A method of treating cancer in a patient, the method comprising:
administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising a radioactive conjugate and one or more pharmaceutically acceptable excipients, wherein:
The radioconjugate comprising at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2,
the radiometal complex comprises a radiometal and
upon administration, the radiometal provides targeted radioactivity of about 50 μci to about 350 μci per dose of the pharmaceutical composition.
2. The method of claim 1, wherein the radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2.
3. The method of claim 2, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequences of SEQ ID No. 1 and SEQ ID No. 2 and SEQ ID No. 3; the light chain variable region comprises the amino acid sequences of SEQ ID NO. 4 and SEQ ID NO. 5 and SEQ ID NO. 6.
4. The method of claim 2 or 3, wherein the antibody comprises a heavy chain variable region (VH) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 9.
5. A method according to claim 2 or 3, wherein the antibody comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID No. 8 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID No. 9.
6. The method of any one of claims 2 to 5, wherein the antibody comprises a heavy chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 10 and a light chain constant region having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 11.
7. The method of any one of claims 2 to 5, wherein the antibody comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID No. 10 and a light chain constant region comprising the amino acid sequence of SEQ ID No. 11.
8. The method according to any one of claims 2 to 7, wherein the antibody comprises a heavy chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 12 and a light chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 13.
9. The method of any one of claims 2 to 7, wherein the antibody comprises a heavy chain having the amino acid sequence of SEQ ID No. 12 and a light chain having the amino acid sequence of SEQ ID No. 13.
10. The method of any one of claims 1 to 9, wherein the radioactive metal is selected from the group consisting of: 225 Ac、 111 In、 177 Lu、 32 P、 47 Sc、 67 Cu、 77 As、 89 Sr、 90 Y、 99 Tc、 105 Rh、 109 Pd、 111 Ag、 131 I、 134 Ce、 149 Tb、 152 Tb、 155 Tb、
153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 255 fm sum 227 Th。
11. The method of any one of claims 1 to 9, wherein the radioactive metal is 225 Ac。
12. The method of any one of claims 1 to 11, wherein the radiometal complex comprises a chelator selected from the group consisting of: 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA), S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA), 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA), 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA), 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A), and derivatives thereof.
13. The method of any one of claims 1 to 11, wherein the radiometal complex comprises a chelator that is DOTA.
14. The method of any one of claims 1 to 13, wherein the radiometal complex comprises a chelate to DOTA 225 Ac。
15. The method of any one of claims 1 to 11, wherein the radioconjugate comprises a radiometal chelated to: (a) A compound of formula (IV)
Or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and is also provided with
R 4 Is an antibody; or alternatively
(b) A compound of formula (V)
Or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and is also provided with
R 4 Is an antibody.
16. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci to about 350 μci per about 2mg total antibody.
17. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 50. Mu. Ci to about 300. Mu. Ci per about 2mg total antibody Sex.
18. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci to about 250 μci per about 2mg total antibody.
19. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci to about 200 μci per about 2mg total antibody.
20. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci to about 150 μci per about 2mg total antibody.
21. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci to about 100 μci per about 2mg total antibody.
22. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 150 μci to about 250 μci per about 2mg total antibody.
23. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 50 μci per about 2mg total antibody.
24. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 100 μci per about 2mg total antibody.
25. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 150 μci per about 2mg total antibody.
26. The method of any one of claims 2 to 15, wherein the radioactive metal is 225 Ac and the radioactive metal provides a targeted specific activity of about 200 μci per about 2mg total antibody.
27. The method of any one of claims 2 to 26, wherein the pharmaceutical composition comprises a targeted radioactivity concentration of about 1 μci/mL to about 100 μci/mL, or about 5 μci/mL to about 75 μci/mL, or about 10 μci/mL to about 60 μci/mL, or about 12.5 μci/mL to about 50 μci/mL, or about 12.5 μci/mL, or about 25 μci/mL, or about 37.5 μci/mL, or about 50 μci/mL.
28. The method of any one of claims 2 to 27, wherein the pharmaceutical composition comprises about 1mg to about 5mg total antibody, or about 1mg to about 4mg total antibody.
29. The method of any one of claims 2 to 27, wherein the pharmaceutical composition comprises about 1mg to about 3mg total antibody.
30. The method of any one of claims 2 to 27, wherein the pharmaceutical composition comprises about 1.5mg to about 2.5mg total antibody.
31. The method of any one of claims 2 to 27, wherein the pharmaceutical composition comprises about 2mg total antibody.
32. The method of any one of claims 1-31, wherein the one or more pharmaceutically acceptable excipients comprise one or more radioprotective agents.
33. The method of claim 32, wherein the one or more radioprotectants comprise sodium ascorbate, gentisic acid or a combination thereof.
34. The method of claim 32, wherein the one or more radioprotectants comprises sodium ascorbate.
35. The method of claim 32, wherein the one or more radioprotectants comprise gentisic acid.
36. The method of any one of claims 1 to 35, wherein the one or more pharmaceutically acceptable excipients further comprise one or more surfactants.
37. The method of claim 36, wherein the one or more surfactants comprise polysorbate 20.
38. The method of any one of claims 1-37, wherein the one or more pharmaceutically acceptable excipients further comprises an acetate buffer.
39. The method of any one of claims 1 to 38, wherein the pharmaceutical composition comprises a radioactive conjugate, sodium ascorbate, polysorbate 20, acetate buffer, and water.
40. The method of any one of claims 1-38, wherein the pharmaceutical composition comprises an aqueous solution of a radioactive conjugate, about 24mM to 28mM acetate, about 0.25% to 0.75% sodium ascorbate, and about 0.01% to 0.1% polysorbate 20.
41. The method of any one of claims 1-38, wherein the pharmaceutical composition comprises an aqueous solution of a radioactive conjugate, about 25mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
42. The method of any one of claims 1-38, wherein the pharmaceutical composition comprises an aqueous solution of a radioactive conjugate, about 26.75mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
43. The method of any one of claims 1 to 42, wherein the pharmaceutical composition has a pH of about 5 to about 6 (e.g., about 5.5).
44. The method of any one of claims 1 to 43, wherein the pharmaceutical composition does not contain any preservative.
45. The method of any one of claims 1 to 44, wherein the pharmaceutical composition does not contain any sucrose.
46. The method of any one of claims 1 to 44, wherein the pharmaceutical composition does not contain any monosaccharides, disaccharides, oligosaccharides or polysaccharides.
47. The method of any one of claims 1 to 44, wherein the pharmaceutical composition does not contain any mono-or disaccharides.
48. The method of any one of claims 1 to 44, wherein the pharmaceutical composition does not contain any disaccharides.
49. The method of any one of claims 1-48, wherein the pharmaceutical composition is stable at a temperature range of about 2 ℃ to 8 ℃ for at least 96 hours or at least 120 hours.
50. The method of any one of claims 2 to 49, wherein the dose of the pharmaceutical composition has a volume of about 1mL to about 20mL, or about 1mL to about 10mL, or about 2mL to about 6mL, or about 3mL to about 5mL, or about 4 mL.
51. The method of any one of claims 2-49, wherein the dose of the pharmaceutical composition comprises about 2mg total antibody per about 4mL dose.
52. The method of any one of claims 2 to 51, wherein the pharmaceutical composition comprises total antibodies in an amount of about 0.01mg/mL to 5.0mg/mL, or about 0.1mg/mL to 1.0mg/mL, or about 0.4mg/mL to 0.6mg/mL, or about 0.5mg/mL (e.g., wherein total antibodies comprise the total amount of conjugate intermediate and radioactive conjugate).
53. The method of any one of claims 2-52, wherein the pharmaceutical composition further comprises a non-radiolabeled antibody, wherein the non-radiolabeled antibody is the same antibody as the antibody conjugated to the radiometal complex.
54. The method of claim 53, wherein the total amount of conjugated antibody and non-radiolabeled antibody is no more than about 10mg, or about 9mg, or about 8mg, or about 7mg, or about 6mg, or about 5mg, or about 4mg, or about 3mg, or about 2mg.
55. The method of any one of claims 1 to 54, comprising administering the pharmaceutical composition intravenously to the patient.
56. The method of any one of claims 1-55, comprising administering the pharmaceutical composition to the patient within about 168 hours, or within about 144 hours, or within about 120 hours, or within about 96 hours, or within about 72 hours, or within about 48 hours, or within about 24 hours after chelating the radiometal to a conjugate intermediate to form the radioconjugate.
57. The method of any one of claims 1-56, comprising administering the pharmaceutical composition once to the patient every about 4 weeks.
58. The method of any one of claims 1-56, comprising administering the pharmaceutical composition to the patient once every about 8 weeks.
59. The method of any one of claims 1-56, comprising administering the pharmaceutical composition to the patient once every about 12 weeks.
60. The method of any one of claims 1 to 59, wherein the cancer is prostate cancer.
61. The method of any one of claims 1 to 59, wherein the cancer is non-localized prostate cancer.
62. The method of any one of claims 1 to 59, wherein the cancer is metastatic prostate cancer.
63. The method of any one of claims 1 to 59, wherein the cancer is castration-resistant prostate cancer (CRPC).
64. The method of any one of claims 1 to 59, wherein the cancer is metastatic castration-resistant prostate cancer (mCRPC).
65. The method of any one of claims 1 to 59, wherein the cancer is mCRPC with adenocarcinoma.
66. The method of any one of claims 1-65, wherein the patient has a testosterone castration level of about 50ng/dL or less.
67. The method of any one of claims 1-66, wherein the patient has been previously exposed to at least one Androgen Receptor (AR) targeted therapy.
68. The method of claim 67, wherein the AR-targeted therapy is abiraterone acetate, enzalutamide, apatamide, darunamine, or a combination of any of the foregoing.
69. The method of any one of claims 1-68, wherein the patient has previously undergone chemotherapy.
70. The method of claim 69, wherein the chemotherapy involves administration of a taxane.
71. The method of any one of claims 1-70, wherein the patient has previously undergone orchiectomy or medical castration.
72. The method of any one of claims 1-71, wherein the patient is receiving androgen ablation therapy with a gonadotropin releasing hormone (GnRH) agonist or antagonist.
73. The method of any one of claims 1-72, comprising administering the dose to the patient in a single administration.
74. The method of any one of claims 1 to 72, comprising administering the dose in multiple administrations of more than one sub-dose.
75. The method of claim 74, comprising administering the dose in two sub-doses.
76. A pharmaceutical composition comprising:
A radio conjugate and one or more pharmaceutically acceptable excipients, wherein:
the radioconjugate comprises at least one radiometal complex conjugated to an antibody or antigen binding fragment having binding specificity for hK2, and
the radiometal complex comprises a radiometal.
77. The pharmaceutical composition of claim 76, wherein the one or more pharmaceutically acceptable excipients comprises one or more radioprotective agents.
78. The pharmaceutical composition according to claim 76 or 77, wherein said radioconjugate comprises at least one radiometal complex conjugated to an antibody having binding specificity for hK 2.
79. The pharmaceutical composition of claim 78, wherein the antibody comprises a heavy chain variable region comprising SEQ ID No. 1 and SEQ ID No. 2 and a light chain variable region
The amino acid sequence of SEQ ID NO. 3; the light chain variable region comprises SEQ ID NO. 4 and
the amino acid sequences of SEQ ID No. 5 and SEQ ID No. 6.
80. The pharmaceutical composition of claim 78 or claim 79, wherein the antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL), the heavy chain variable region being identical to SEQ id no
The amino acid sequence of ID No. 8 has at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity, and the light chain variable region has at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 9.
81. The pharmaceutical composition of claim 78 or claim 79, wherein the antibody comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID No. 8 and a heavy chain variable region (VH) comprising SEQ ID No. 8
The light chain variable region (VL) of the amino acid sequence of ID No. 9.
82. The pharmaceutical composition of any one of claims 78 to 81, wherein the antibody comprises a heavy chain constant region having at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% of the amino acid sequence of SEQ ID No. 10 and a light chain constant region
Sequence identity, the light chain constant region has at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID NO. 11.
83. The pharmaceutical composition of any one of claims 78 to 81, wherein the antibody comprises a heavy chain constant region comprising the amino acid sequence of SEQ ID No. 10 and a light chain constant region comprising SEQ ID No. 10
The light chain constant region of the amino acid sequence of NO. 11.
84. The pharmaceutical composition of any one of claims 78 to 83, wherein the antibody comprises a heavy chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 12 and a light chain having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% sequence identity to the amino acid sequence of SEQ ID No. 13.
85. The pharmaceutical composition of any one of claims 78 to 83, wherein said antibody comprises a heavy chain having the amino acid sequence of SEQ ID No. 12 and a light chain having the amino acid sequence of SEQ ID No. 13.
86. The pharmaceutical composition of any one of claims 77-85, wherein said radioactive metal is selected from the group consisting of: 225 Ac、 111 In、 177 Lu、 32 P、 47 Sc、 67 Cu、
77 As、 89 Sr、 90 Y、 99 Tc、 105 Rh、 109 Pd、 111 Ag、 131 I、 134 Ce、 149 Tb、 152 Tb、 155 Tb、 153 Sm、 159 Gd、 165 Dy、 166 Ho、 169 Er、 186 Re、 188 Re、 194 Ir、 198 Au、 199 Au、 211 At、 212 Pb、 212 Bi、 213 Bi、 223 Ra、 255 fm sum 227 Th。
87. The pharmaceutical composition of any one of claims 77-85, wherein the radiometal is 225 Ac。
88. The pharmaceutical composition of any one of claims 77-87, wherein the radiometal complex comprises a chelator selected from the group consisting of: 1,4,7, 10-tetraazacyclododecane-1, 4,7, 10-tetraacetic acid (DOTA), S-2- (4-isothiocyanatobenzyl) -1,4, 7-triazacyclononane-1, 4, 7-triacetic acid (NOTA), 1,4,8, 11-tetraazacyclododecane-1, 4,8, 11-tetraacetic acid (TETA), 3,6,9,15-tetraazabicyclo [9.3.1] -pentadecane-1 (15), 11, 13-triene-4- (S) - (4-isothiocyanatobenzyl) -3,6, 9-triacetic acid (PCTA), 5-S- (4-aminobenzyl) -1-oxa-4, 7, 10-triazacyclododecane-4, 7, 10-tris (acetic acid) (DO 3A), and derivatives thereof.
89. The pharmaceutical composition of any one of claims 77-87, wherein the radiometal complex comprises a chelator that is DOTA.
90. The pharmaceutical composition of any one of claims 77-89, wherein the radiometal complex comprises a chelate to DOTA 225 Ac。
91. The pharmaceutical composition of any one of claims 77-87, wherein the radioconjugate comprises a radiometal chelated to: (a) A compound of formula (IV)
Or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen and R 2 is-L 1 -R 4
Alternatively, R 1 is-L 1 -R 4 And R is 2 Is hydrogen;
R 3 is hydrogen;
alternatively, R 2 And R is 3 Taken together with the carbon atoms to which they are attached to form a 5-or 6-membered cycloalkyl group, wherein the 5-or 6-membered cycloalkyl group is optionally substituted with-L 1 -R 4 Substitution;
L 1 absent or a linker; and is also provided with
R 4 Is an antibody; or alternatively
(b) A compound of formula (V)
Or a pharmaceutically acceptable salt thereof, wherein:
L 1 absent or a linker; and is also provided with
R 4 Is an antibody.
92. The pharmaceutical composition of any one of claims 77-91, wherein the one or more radioprotectants comprise sodium ascorbate, gentisic acid, or a combination thereof (e.g., in an amount of about 0.1w/v% to 1w/v%, or about 0.25w/v% to 0.75w/v%, or about 0.5 w/v%).
93. The pharmaceutical composition of any one of claims 77-91, wherein the one or more radioprotectants comprise sodium ascorbate (e.g., in an amount of about 0.1w/v% to 1w/v%, or about 0.25w/v% to 0.75w/v%, or about 0.5 w/v%).
94. The pharmaceutical composition of any one of claims 77-91, wherein the one or more radioprotectants comprise gentisic acid (e.g., in an amount of about 0.1w/v% to 1w/v%, or about 0.25w/v% to 0.75w/v%, or about 0.5 w/v%).
95. The pharmaceutical composition of any one of claims 76-94, wherein the one or more pharmaceutically acceptable excipients further comprises one or more surfactants.
96. The pharmaceutical composition of claim 95, wherein the one or more surfactants comprise polysorbate 20.
97. The pharmaceutical composition of any one of claims 76-96, wherein the one or more pharmaceutically acceptable excipients further comprises an acetate buffer.
98. The pharmaceutical composition of any one of claims 76-97, comprising a radioactive conjugate, sodium ascorbate, polysorbate 20, acetate buffer, and water.
99. The pharmaceutical composition of any one of claims 76-97, comprising an aqueous solution of a radioactive conjugate, about 24mM to 28mM acetate, about 0.25% to 0.75% sodium ascorbate, and about 0.01% to 0.1% polysorbate 20.
100. The pharmaceutical composition of any one of claims 76-97, comprising an aqueous solution of a radioactive conjugate, about 26.75mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
101. The pharmaceutical composition of any one of claims 76-97, comprising an aqueous solution of a radioactive conjugate, about 26.75mM acetate, about 0.5% sodium ascorbate, and about 0.04% polysorbate 20.
102. The pharmaceutical composition of any one of claims 76-101, wherein the pharmaceutical composition has a pH of about 5 to about 6 (e.g., about 5.5).
103. The pharmaceutical composition of any one of claims 76-101, wherein the pharmaceutical composition is free of any preservative.
104. The pharmaceutical composition of any one of claims 76-103, wherein the pharmaceutical composition does not contain any sucrose.
105. The pharmaceutical composition of any one of claims 76-104, wherein the pharmaceutical composition does not contain any monosaccharides, disaccharides, oligosaccharides or polysaccharides.
106. The pharmaceutical composition of any one of claims 76-104, wherein the pharmaceutical composition does not contain any mono-or disaccharides.
107. The pharmaceutical composition of any one of claims 76-104, wherein the pharmaceutical composition does not contain any disaccharides.
108. The pharmaceutical composition of any one of claims 76-104, wherein the one or more pharmaceutically acceptable excipients consist of or consist essentially of an acetate buffer, sodium ascorbate, and an aqueous solution of polysorbate 20.
109. The pharmaceutical composition of any one of claims 76-108, wherein the pharmaceutical composition is formulated for intravenous administration.
110. The pharmaceutical composition of any one of claims 76-109, wherein the pharmaceutical composition is stable at a temperature range of about 2 ℃ to 8 ℃ for at least 72 hours, or at least 96 hours, or at least 120 hours.
111. The pharmaceutical composition of any one of claims 77-110, wherein said radioactive conjugate comprises an average of about 1 to about 4, or about 2 to about 3, chelator molecules conjugated to said antibody.
112. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 350 μci per about 2mg total antibody.
113. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 300 μci per about 2mg total antibody.
114. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 250 μci per about 2mg total antibody.
115. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 200 μci per about 2mg total antibody.
116. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 150 μci per about 2mg total antibody.
117. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radioactive metal provides a specific activity of about 50 μci to about 100 μci per about 2mg total antibody.
118. The pharmaceutical composition according to any one of claims 77 to 111,wherein the radioactive metal is 225 Ac and the radiometal provides a targeted specific activity of about 50 μci to about 200 μci per about 2mg total antibody when administered.
119. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 50 μci per about 2mg total antibody when administered.
120. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 100 μci per about 2mg total antibody when administered.
121. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 150 μci per about 2mg total antibody when administered.
122. The pharmaceutical composition of any one of claims 77-111, wherein the radiometal is 225 Ac and the radiometal provides a targeted specific activity of about 200 μci per about 2mg total antibody when administered.
123. The pharmaceutical composition of any one of claims 77-122, comprising about 1mg to about 20mg total antibody.
124. The pharmaceutical composition of any one of claims 77-122, comprising about 1mg to about 10mg total antibody.
125. The pharmaceutical composition of any one of claims 77-122, comprising about 1mg to about 5mg total antibody.
126. The pharmaceutical composition of any one of claims 77-122, comprising about 2mg total antibody.
127. The pharmaceutical composition of any one of claims 77-122, comprising about 10mg of total antibody.
128. The pharmaceutical composition of any one of claims 77-127, comprising a total amount of conjugate intermediate and the radioactive conjugate of about 0.1mg/mL to 1.0 mg/mL.
129. The pharmaceutical composition of any one of claims 77-127, comprising a total amount of conjugate intermediate and the radioactive conjugate of about 0.4mg/mL to 0.6 mg/mL.
130. The pharmaceutical composition of any one of claims 77-127, comprising a total amount of about 0.5mg/mL of conjugate intermediate and the radioactive conjugate.
131. The pharmaceutical composition of any one of claims 77-130, further comprising a non-radiolabeled antibody, wherein the non-radiolabeled antibody is the same antibody as the antibody conjugated to the radiometal complex.
132. The pharmaceutical composition of claim 131, wherein the total amount of conjugated antibody and non-radiolabeled antibody is no more than about 10mg, or about 9mg, or about 8mg, or about 7mg, or about 6mg, or about 5mg, or about 4mg, or about 3mg, or about 2mg.
133. A method for treating cancer in a patient, the method comprising administering to the patient a therapeutically effective amount of the pharmaceutical composition of any one of claims 76-132.
134. The method of claim 133, comprising administering the pharmaceutical composition once to the patient every about 4 weeks.
135. The method of claim 133, comprising administering the pharmaceutical composition to the patient once every about 8 weeks.
136. The method of claim 133, comprising administering the pharmaceutical composition to the patient once every about 12 weeks.
137. The method according to any one of claims 133-136, wherein the cancer is prostate cancer.
138. The method of any one of claims 133-136, wherein the cancer is non-localized prostate cancer.
139. The method according to any one of claims 133-136, wherein the cancer is metastatic prostate cancer.
140. The method of any one of claims 133-136, wherein the cancer is castration-resistant prostate cancer (CRPC).
141. The method of any one of claims 133-136, wherein the cancer is metastatic castration-resistant prostate cancer (mCRPC).
142. The method of any one of claims 133-136, wherein the cancer is mCRPC with adenocarcinoma.
143. The method according to any one of claims 133-142, wherein the patient has a testosterone castration level of about 50ng/dL or less.
144. The method of any one of claims 133-142, wherein the patient has been previously exposed to at least one Androgen Receptor (AR) targeted therapy.
145. The method of claim 144, wherein the AR-targeted therapy is abiraterone acetate, enzalutamide, apatamide, darunamine, or a combination of any of the foregoing.
146. The method of any one of claims 133-145 wherein the patient has previously undergone chemotherapy.
147. The method of claim 146, wherein the chemotherapy involves administration of a taxane.
148. The method of any one of claims 133-147, wherein the patient has previously undergone orchiectomy or medical castration.
149. The method of any one of claims 133-148 wherein the patient is receiving androgen ablation therapy with a gonadotropin releasing hormone (GnRH) agonist or antagonist.
CN202280037818.3A 2021-05-27 2022-05-25 Compositions and methods for treating prostate cancer Pending CN117396234A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/193704 2021-05-27
US202263335761P 2022-04-28 2022-04-28
US63/335761 2022-04-28
PCT/IB2022/054891 WO2022249089A1 (en) 2021-05-27 2022-05-25 Compositions and methods for the treatment of prostate cancer

Publications (1)

Publication Number Publication Date
CN117396234A true CN117396234A (en) 2024-01-12

Family

ID=89437766

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280037818.3A Pending CN117396234A (en) 2021-05-27 2022-05-25 Compositions and methods for treating prostate cancer

Country Status (1)

Country Link
CN (1) CN117396234A (en)

Similar Documents

Publication Publication Date Title
JP7487250B2 (en) Antigen-binding constructs for target molecules
JP4354280B2 (en) RS7 antibody
JP4663020B2 (en) New methods for diagnosis and treatment of tumors
EP1521775B1 (en) Monoclonal antibody pam4 and its use for diagnosis and therapy of pancreatic cancer
BR112020012099A2 (en) polypeptide radiolabeling
US20210017295A1 (en) Bispecific binding agents and uses thereof
KR20210046016A (en) Anti-tissue factor antibody-drug conjugates and their use in cancer treatment
US20230011134A1 (en) Compositions and methods for the treatment of prostate cancer
CN117396234A (en) Compositions and methods for treating prostate cancer
EP4317188A1 (en) Radioactive complex of anti-egfr antibody, and radiopharmaceutical
TW202400162A (en) Radiopharmaceutical complexes and combinations
AU2021361746A1 (en) Radioactive complexes of anti-her2 antibody, and radiopharmaceutical
JP2024519970A (en) Trivalent radioisotope biologically targeted radiopharmaceuticals, methods of preparation and uses
WO2023056474A1 (en) Mouse anti-human monoclonal antibody against glypican-3
CN117377690A (en) Radioactive complexes and radiopharmaceuticals of anti-EGFR antibodies
EA045797B1 (en) RADIOACTIVE LABELING OF POLYPEPTIDES
IL165752A (en) Chimeric monoclonal antibody pam4, a conjugate comprising it and an antibody comprising the same or fragments thereof
KR20100077052A (en) Conjugates comprising an antibody specific for the ed-b domain of fibronectin and their use for the detection and treatment of tumours
WO2011110490A1 (en) Process for the production of radioactively labelled scfv antibody fragments, kits and compositions

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