The instant application contains a Sequence Listing (ABV21624USL2_ST26.xml; Size: 27,908 bytes; and Date of Creation: Sep. 27, 2024), which has been submitted electronically and is hereby incorporated by reference in its entirety.
Prostate cancer is the second most diagnosed cancer and the fifth leading cause of cancer death among men worldwide. Currently there are no curative therapies available for metastatic prostate cancer.
PSMA (prostate specific membrane antigen) and STEAP1 (six transmembrane epithelial antigen of the prostate-1) are well characterized antigens overexpressed in metastatic prostate cancer with low expression in normal tissue.
PSMA plays a role in the metabolism of folates that may contribute to prostate cancer progression. PSMA expression is increased in metastatic and hormone refractory tumors and correlates with tumor aggressiveness, stage, recurrence, and castration resistance.
STEAP1 is an ion channel or transporter that modulates the concentration of small molecules, ions and nutrients. STEAP1 functions in cellular communication and in cell adhesion processes. STEAP1 is overexpressed in advanced and metastatic tumors, where it is associated with aggressive cell proliferation and invasion.
Antibody drug conjugates (ADCs) are a rapidly growing class of cancer drugs that combine the cell surface targeting properties of antibodies conjugated to potent cytotoxic drugs using chemical linkers. An advantage of ADCs is their ability to deliver toxic payloads to a tumor, reducing toxicity associated with systemic exposure to normal tissues.
There is a need for novel therapeutics combining binding agents of prostate cancer antigens to enhance tumor coverage, mitigate resistance, and drive durable response.
In some aspects, the present disclosure provides a dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
In some embodiments, the first heavy chain variable domain (VH1) comprises the amino acid sequence of SEQ ID NO: 2; the second heavy chain variable domain (VH2) comprises the amino acid sequence of SEQ ID NO: 7; the first light chain variable domain (VL1) comprises the amino acid sequence of SEQ ID NO: 12; and the second light chain variable domain (VL2) comprises the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the first peptide linker (L1) comprises the amino acid sequence GGGGSGGGGS (SEQ ID NO: 6). In some embodiments, the second peptide linker (L2) comprises the amino acid sequence GGSGGGGSG (SEQ ID NO: 16). In some embodiments, the IgG1 heavy chain constant region (CH1CH2CH3) comprises the amino acid sequence of SEQ ID NO: 21 or 23. In some embodiments, the kappa light chain constant region (Cκ) comprises the amino acid sequence of SEQ ID NO: 22.
In some embodiments, each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24; and each light chain comprises the amino acid sequence of SEQ ID NO: 11.
In some embodiments, the first heavy chain and the second heavy chain are connected by two disulfide bridges, and the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering.
In some embodiments, the Top1i linker-Drugs are conjugated to the cysteines at position C220 of the first and the second heavy chains of the DVD-Ig according to EU numbering.
In some embodiments, n is 1. In some embodiments, n is 2.
In some aspects, the present disclosure provides a dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
In some aspects, described herein are novel DVD-Ig molecules that bind to both human STEAP1 and human PSMA, as well as drug conjugates of the said DVD-Ig molecules.
PSMA and STEAP1 are well characterized antigens overexpressed in metastatic prostate cancer with low expression in normal tissue. Both PSMA and STEAP1 are expressed over a range of intensities within prostate cancer cells. Such intratumor heterogeneity can hinder successful treatment of prostate cancer. In the present disclosure, the DVD-Ig platform is utilized for dual targeting of PSMA and STEAP1, which increases both breath and depth of responses relative to monospecific ADCs. Compared to monospecific ADCs, which target a single antigen PSMA or STEAP1, targeting both antigens offers several advantages: (1) dual targeting has broader efficacy on tumor cells and captures larger patient populations; (2) DVD-Ig molecules conjugated with cytotoxic drugs (DVD-Ig DCs) show enhanced internalization and activity than either monospecific ADC; and (3) compared to administration of a combination of two monospecific ADCs, DVD-Ig DCs reduce total dosing of agent(s) required.
DVD-Ig are symmetrical molecules with two identical light chains and two identical heavy chains and therefore have a more simplified production process than four-chain asymmetric formats that can suffer from chain mis-pairing and require downstream purification. However, identification of a DVD-Ig that is worthy of being a drug candidate, with the desired potency, physicochemical properties, efficacy, and manufacturability is challenging. The first challenge inherent to its design, is that the inner variable region can have reduced or eliminated binding affinity to its target. This is due to steric hindrance and can sometimes be mitigated by the choice of linker between the variable regions. A second challenge is that the expression level of some DVD-Igs can be lower than that of the parental IgG from which they are derived. Thirdly, the drug-like properties such as thermal stability, aggregation, or ability to achieve the high concentrations required for drug product formulation can be poor for some molecules. DVD-Igs described in the present disclosure have good expression level and strong binding specificity directed towards both human STEAP and human PSMA, and potent induction of cancer cell killing.
To obtain the DVD-Igs, 200 molecules were designed and cloned for expression. 149 molecules were successfully expressed, and 122 molecules had sufficient yield for characterization. 45 molecules had dual binding activity, and 20 were selected for additional characterization. A lead molecule was identified for further development, as it demonstrated good cell killing potency and acceptable drug-like properties.
The singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.
The term “and/or” as used in a phrase such as “A and/or B” herein is intended to mean “A and B”, “A or B”, “A” or “B”.
“Binding to human STEAP1 or human PSMA” refers to a molecule that is capable of binding human STEAP1 or human PSMA with sufficient affinity such that the molecule is useful as a therapeutic agent in targeting human STEAP or human PSMA.
The term “CDR” refers to a complementarity determining region within an immunoglobulin variable region sequence. There are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3. The term “CDR set” refers to a group of three CDRs that are present in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al. (1987) Sequences of Proteins of Immunological Interest, Fourth Edition. US Govt. Printing Off. No. 165-492; and Kabat et al. (1991) Sequences of Proteins of Inimunological Interest, Fifth Edition. NIH Publication No. 91-3242) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody or binding protein, but also provides precise residue boundaries defining the three CDRs in each heavy or light chain sequence. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia and Lesk (1987) J. Mol. Biol. 196:901-917; Chothia et al. (1989) Nature 342:877-883) found that certain sub-portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub-portions were designated as L1, L2 and L3 or H1, H2 and H3 where the “L” and the “H” designates the light chain and the heavy chain regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described in Padlan (1995) FASEB J. 9:133-139 and MacCallum (1996) J. Mol. Biol. 262(5):732-45). Still other CDR boundary definitions may not strictly follow one of the herein described systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. In some embodiments, the CDRs of the DVD-Igs of the present disclosure are defined by EU numbering.
The terms “Kabat numbering”, “Kabat definition” and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody or binding protein, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad. Sci. 190:382-391 and, Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50-56 for CDR2, and amino acid positions 89 to 97 for CDR3.
In some embodiments, the constant region (heavy chain constant region and/or light chain constant region) sequences are identified using EU numbering. The term “EU numbering”, which is recognized in the art, refers to a system of numbering amino acid residues based on Edelman, G. M. et al., Proc. Natl. Acad. USA, 63, 78-85 (1969). The numbering schemes disclosed herein, including EU numbering and Kabat numbering, are based on the conventional antibody structure. Because DVD-Ig has an extra variable domain (i.e., outer variable domain) connected to the N-terminal of the inner variable domain via a flexible linker, one of ordinary skill in the art would understand that the exact position of the amino acid based on the DVD-Ig structure would be different from the EU numbering or Kabat numbering. However, one of ordinary skill in the art would readily identify the corresponding amino acid within the DVD-Ig sequence based on the number schemes (e.g., EU numbering and Kabat numbering) disclosed herein. One of skill in the art could also identify positions of the DVD-Ig amino acid sequence by counting from the processed N-terminus, which does not conform to any numbering scheme.
The term “Fc” or “Fc region” is a term well known to the skilled artisan and is involved in complement activation, Clq binding, C3 activation and Fc receptor binding.
The term “hinge region” refers to a flexible amino acid stretch in the central part of the heavy chains of immunoglobulin antibodies, which links these 2 chains by disulfide bonds. The “variable domain” or “variable region” (variable domain/region of a light chain (VL), variable domain/region of a heavy chain (VH)) as used herein denotes each of the pair of light and heavy chains which are involved directly in binding the antibody to the target. The domains of variable human light and heavy chains have the same general structure and each domain comprises at least one complementary determining region (CDR), preferably three CDRs, which play a particularly important role in the binding specificity/affinity of the antibodies.
The dual variable domain immunoglobulin (DVD-Ig) combines the target binding domains of two monoclonal antibodies (mAbs) via a flexible linker, which yields a tetravalent IgG-like molecule. DVD-Ig is symmetrical, bispecific antibody-like biologics with a 2+2 antigen binding stoichiometry. In some embodiments, in a linear representation of the mature sequence, the DID-Ig comprises an outer variable domain (VH1 or VL1) connected to the inner variable domain (VH2 or VL2) via a linker (L), with a standard Heavy Chain (HC) or Light Chain (LC) constant region at the C-terminus.
In some embodiments, the DVD-Ig provided herein comprises outer variable domains (VH1 and VL1) that bind human STEAP1 and inner variable domains (VH2 and VL2) that bind human PSMA. The diagram depicting the design of DVD-Ig molecule, according to an embodiment of the invention, is shown in
For example, in some aspects, the present disclosure provides a DVD-Ig that binds human STEAP1 and human PSMA comprising two identical heavy chains and two identical light chains,
In some embodiments, the first heavy chain variable domain (VH1) comprises the amino acid sequence of SEQ ID NO: 2; the second heavy chain variable domain (VH2) comprises the amino acid sequence of SEQ ID NO: 7; the first light chain variable domain (VL1) comprises the amino acid sequence of SEQ ID NO: 12; and the second light chain variable domain (VL2) comprises the amino acid sequence of SEQ ID NO: 17.
In some embodiments, each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24; and each light chain comprises the amino acid sequence of SEQ ID NO: 11.
Flexible Glycine-Serine rich linkers are preferred for DVD-Igs. The GS linkers can be for DVD-Igs of the present disclosure include, but are not limited to, the ones listed in the table below. In some embodiments, the linker (e.g., the linker connecting VH1 and VH2, or L1) comprises GGGGSGGGGS (SEQ ID NO: 6). In some embodiments, the linker (e.g., the linker connecting VL1 and VL2, or L2) comprises GGSGGGGSG (SEQ ID NO: 16).
In some embodiments, the kappa light chain constant region (Cκ) is a human Cκ or is derived from a human Cκ region. In some embodiments, the LC cysteine that normally forms the interchain disulfide bond with C220 of the heavy chain (HC) has been mutated to alanine, which creates an unpaired cysteine at position C220 of the HC for conjugation of the linker drug in a facile, site-specific manner. For example, in some embodiments, the kappa light chain constant region (Cκ) is derived from the human Cκ and comprises an amino acid substitution of C214A according to EU numbering of the amino acid number of the antibody chain.
In some embodiments, the kappa light chain constant region (Cκ) comprises the amino acid sequence of SEQ ID NO: 22.
In one embodiment, CH2CH3 of the first and the second heavy chains form a dimeric Fc region. In one embodiment, the Fc region of the present invention is derived from human origin. In one embodiment, the Fc region of the present invention is a human IgG1 Fc region or derived from a human IgG1 Fc region.
In some embodiments, the Fc region comprises a first CH2CH3 region and a second CH2CH3 region, wherein the first CH2CH3 region is covalently bound by two disulfide bridges to the second CH2CH3 region. In some embodiments, the first heavy chain and the second heavy chain are connected by two disulfide bridges, and the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering. For example, in some embodiments, the first heavy chain and the second heavy chain have an amino acid sequence of SEQ ID NO: 1 or 24, and the two disulfide bridges are between C362 of the first heavy chain of SEQ ID NO: 1 or 24 and C362 of the second heavy chain of SEQ ID NO: 1 or 24, and between C365 of the first heavy chain of SEQ ID NO: 1 or 24 and C365 of the second heavy chain of SEQ ID NO: 1 or 24.
In one embodiment, the Fc region of the present invention comprises a hinge region. Various hinge regions can be used in the bispecific molecules of the present invention, for example, to optimize certain characteristics. In an illustrative example, one or more amino acid substitutions, insertions, and/or deletions within a hinge region of a human IgG1, IgG2, IgG3 or IgG4 can be introduced to reduce the level or rate of fragmentation and/or aggregation.
In one embodiment, the Fc region of the present invention comprises one or more mutations to modulate (e.g., reduce) IgG Fc (Fragment crystallizable) receptor and complement binding. In one embodiment, the Fc region of the present invention comprises one or more mutations to modulate (e.g., reduce) Fc receptor-based function of the Fc region. In one embodiment, the Fc region of the present invention comprises one or more mutations to modulate (e.g., reduce) FcγR-based effector function of the Fc region. In one embodiment, the Fe region of the present invention is derived from the human IgG1 Fe region and comprises an amino acid substitution of L234A and/or L235A according to EU numbering of the amino acid number of the antibody chain.
In one embodiment, the CH1CH2CH3 of the present invention comprises the amino acid sequence of SEQ ID NO: 21 or 23.
In some embodiments, the polypeptide sequence of either or both heavy chains of the DVD-Ig molecules described herein lack the C-terminal lysine, resulting in a C-terminal glycine residue. For example, in some embodiments, either or both heavy chains of the DVD-Ig molecules described herein comprise the amino acid sequence of SEQ ID NO: 24.
In some aspects, the present disclosure provides a DVD-Ig that binds human STEAP1 and human PSMA comprising two identical heavy chains and two identical light chains, wherein each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24, and each light chain comprises the amino acid sequence of SEQ ID NO: 11; and wherein the first heavy chain and the second heavy chain are connected by two disulfide bridges, and wherein the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering.
In some embodiments, DVD-Ig molecules described herein are conjugated to a drug, e.g., a Topoisomerase 1 Inhibitor (TOP1i). Topoisomerase 1 (TOP1) removes supercoils formed during DNA replication. TOP1 inhibitors (TOP1i) can bind and stabilize TOP1-DNA complexes, inducing DNA strand breakage and apoptosis. In some embodiments, DVD-Ig molecules described herein are conjugated to a Topoisomerase 1 inhibitor drug (“TOP1i drug) according to the structural formula (I).
In some embodiments, a TOP1i drug is a compound according to formula (I). In embodiments, the TOP1i drug is (7S)-14-(3-aminobicyclo [1.1.1]pentan-1-yl)-7-ethyl-7-hydroxy-2H, 10H-[1,3]dioxolo[4,5-g]pyrano[3′,4′: 6,7]indolizino[1,2-b]quinoline-8,11 (7H,13H)-dione.
TOP1i drugs as contemplated herein may be conjugated to a DVD-Ig described herein as DVD-Ig DC as shown in structural formula (II):
wherein represents the point of attachment of a linker to the TOP1i drug.
Topoisomerase inhibitors as described herein may be conjugated to DVD-Ig described herein to form a DVD-Ig TOP1i drug conjugate. DVD-Ig-drug conjugates may increase the therapeutic efficacy of DVD-Ig in treating disease due to the ability of the DVD-Ig DC to selectively deliver one or more drug moiety(s) to target tissues, such as a tumor-associated antigen, e.g., human STEAP1 and/or human PSMA expressing tumors. Thus, in embodiments, the present disclosure provides DVD-Ig DCs described herein for therapeutic use, e.g., in the treatment of prostate cancer.
In certain embodiments, a TOP1i drug is conjugated to an antibody by way of linker according to structural formula (III):
wherein X represents bromo or an N-linked maleimide.
In some embodiments of the DVD-Ig DCs described herein, TOP1i drugs are conjugated to the DVD-Igs by way of a linker moiety. As will be appreciated by skilled artisans, the linkers connect the TOP1i drug to the DVD-Ig by forming a covalent linkage to the TOP1i drug at one location and a covalent linkage to the antibody at another. The covalent linkages are formed by reaction between functional groups on the linker and functional groups on the TOP1i drug and the DVD-Ig. In some embodiments, the Top1i linker-Drugs are conjugated to the cysteines at position C220 of the first and the second heavy chains of the DVD-Ig described herein.
In some embodiments, the synthetic intermediate compounds that may be used to form DVD-Igs may include the Linker Drug (LD) shown as in Structural Formula (V))
(2S)-2-(2-bromoacetamido)-N-[(2S)-1-({3-[(7S)-7-ethyl-7-hydroxy-8,11-dioxo-7,8,11,13-tetrahydro-2H,10H-[1,3]dioxolo[4,5-g]pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-14-yl]bicyclo[1[0.1.1]pentan-1-yl}amino)-1-oxopropan-2-yl]-3-methylbutanamide
The DVD-Ig DCs disclosed herein comprise drug molecules linked to DVD-Ig moieties in various stoichiometric molar ratios depending on the configuration of the DVD-Ig and, at least in part, the method used to effect conjugation.
The terms “drug load” or “drug loading” refer to the number of drug molecules per antibody in an individual DVD-Ig DC molecule. The number of TOP1i drugs linked to an DVD-Ig DC can vary and will be limited by the number of available attachments sites on the DVD-Ig. As contemplated for the DVD-Ig DCs of the invention, the linker will link a single TOP1i drug to a DVD-Ig. In some embodiments, the DVD-Ig DCs described herein have an n of 1 or 2. In embodiments, n is 1. In embodiments, n is 2. In embodiments, the drug loading may comprise 1 drug molecule, or 2 drug molecules.
In some aspects, the present disclosure provides a dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
In some embodiments, the first heavy chain variable domain (VH1) comprises the amino acid sequence of SEQ ID NO: 2; the second heavy chain variable domain (VH2) comprises the amino acid sequence of SEQ ID NO: 7; the first light chain variable domain (VL1) comprises the amino acid sequence of SEQ ID NO: 12; and the second light chain variable domain (VL2) comprises the amino acid sequence of SEQ ID NO: 17.
In some embodiments, each heavy chain comprises the amino acid sequence of SEQ ID NO: 1 or 24; and each light chain comprises the amino acid sequence of SEQ ID NO: 11.
In some embodiments, the first peptide linker (L1) comprises the amino acid sequence GGGGSGGGGS (SEQ ID NO: 6). In some embodiments, the second peptide linker (L2) comprises the amino acid sequence GGSGGGGSG (SEQ ID NO: 16).
In some embodiments, the IgG1 heavy chain constant region (CH1CH2CH3) comprises the amino acid sequence of SEQ ID NO: 21 or 23. In some embodiments, the kappa light chain constant region (Cκ) comprises the amino acid sequence of SEQ ID NO: 22.
In some embodiments, the first heavy chain and the second heavy chain are connected by two disulfide bridges, and the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain, wherein the amino acid positions are numbered using EU numbering. For example, in some embodiments, the first heavy chain and the second heavy chain have an amino acid sequence of SEQ ID NO: 1 or 24, and the two disulfide bridges are between C362 of the first heavy chain of SEQ ID NO: 1 or 24 and C362 of the second heavy chain of SEQ ID NO: 1 or 24, and between C365 of the first heavy chain of SEQ ID NO: 1 or 24 and C365 of the second heavy chain of SEQ ID NO: 1 or 24.
In some embodiments, the Top1i linker-Drugs are conjugated to the cysteines at position C220 of the first and the second heavy chains of the DVD-Ig according to EU numbering.
In some embodiments, n is 1.
In some embodiments, n is 2.
In some aspects, the present disclosure provides a dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
In another aspect, the present disclosure pertains to a pharmaceutical composition comprising a DVD-Ig molecule or a DVD-Ig DC described herein and a pharmaceutically acceptable excipient or carrier.
In another aspect, the present disclosure pertains to a method of treating prostate cancer, comprising administering to a subject (e.g., a patient) in need thereof, a DVD-Ig drug conjugate described herein, or a pharmaceutical composition thereof.
In another aspect, the present disclosure pertains to nucleic acid molecules encoding a heavy chain and/or a light chain of a DVD-Ig molecule described herein.
In another aspect, the present disclosure pertains to vectors comprising nucleic acid molecules encoding a heavy chain and/or a light chain of a DVD-Ig molecule described herein.
In another aspect, the present disclosure pertains to host cells capable of producing a DVD-Ig molecule described herein.
The following Examples are provided for purposes of illustration, and not limitation.
PSMA and STEAP1 are well characterized antigens overexpressed in metastatic prostate cancer with low expression in normal tissue. Both PSMA and STEAP1 are expressed at varying levels in most prostate tumors. To bind both antigens on tumor cells with varied antigen expression, DVD-Igs that target PSMA and STEAP1 were generated. DVD-Ig that targets both antigens can capture a larger patient population, as it can be effective in patients with PSMA1 “high” tumors, STEAP1 “high” tumors, and dual “moderate” expressors (
To mitigate risks inherent in the DVD-Ig format, multiple variable regions for PSMA and STEAP1 were identified and humanized for inclusion in a large panel of molecules for screening. The variable regions were derived from mAbs with favorable expression and drug-like property profiles, as any weakness in a variable domain could be amplified when incorporated into a bispecific format. Notably, two well-behaved variable regions could have interactions with each other when placed in a DVD-Ig format that subsequently causes unfavorable properties. Therefore, a library of candidate DVD-Igs were designed and constructed, and a screening was carried out to identify DVD-Igs that have preferred properties.
Parental mAbs for the DVD-Ig molecule were generated using mouse (PSMA) and rat (STEAP1) hybridoma technology and hybridoma supernatants were subsequently screened for activity. Antibody heavy and light chain variable regions (VH and VL) of the selected hybridomas were cloned and expressed as chimeric antibodies, and then further humanized using computer-aided high throughput humanization design software developed internally at AbbVie. Each of the 10 selected humanized PSMA variable regions and each of the 10 selected humanized STEAP1 variable regions were combined with a linker (GS10 on the HC, GS9 on the LC) in both orientations, which resulted in a target library of 200 DVD-Ig molecules.
In transient mammalian expression systems, DVD-Igs were expressed with two plasmids, one for the HC and one for the LC. The plasmids were constructed by overlap extension PCR and ligation. Briefly, each variable domain was amplified by PCR with primers to incorporate an appropriate overhang to the vector or linker. The PCR products were purified and combined for overlap extension PCR. Cleaned up inserts and the pHybE cloning vectors with the constant region were then digested with EcoRI/SalI for the heavy chain (HC) and EcoRI/BsiWI for the light chain (LC). Pooled ligation reactions were transformed into DH5 alpha cells and a 6× oversampling of colonies was screened by colony PCR. For pools with low recovery this step was repeated.
For all DVD-Ig for which matching sequences were identified for both HC and LC plasmids, bacterial cell cultures were re-arrayed into 96-well plates and plasmid DNA was purified for transfection.
The 5 mL scale HTAP platform was used to express and purify the DVD-Ig in Expi293 cells (Thermo). This production scale generally yields about 100 mg of protein for screening. A Hamilton STAR robot was used to transfect DNA from Costar 3357 96-well plates containing HC and LC DNA in four 24-well plates at a 3:2 ratio with ExpiFectamine™ (Gibco A29129). Cultures were fed on day 1. After 5-day transfection, the supernatants containing the DVD-Ig were harvested, re-arrayed and filtered for purification. Protein A purification was done in the 96-well plate format on the Assaymap Bravo. Purified proteins were screened for binding to both PSMA and STEAP1, internalization, and stability by Size Exclusion Chromatography (SEC).
149 of 200 DVD-Igs were successfully expressed, and 122 DVD-Igs had sufficient yield for full characterization. Forty-five dual target binders were identified by FACS binding assay, and 20 were selected for further characterization (
The initial drug like properties including the aggregation profiles and the thermal stability of the DVDs shows that a significant percentage of DVD-Igs produced do not meet the aggregation threshold of at least 90% monomer or DSF thermal stability of T-onset >55° C. threshold.
An indirect cytotoxicity assay was used to characterize the HTAP DVD-Igs. LNCaP.FGC cells cultured in RPMI media supplemented with 10% heat-inactivated FBS, and HEK293 cells engineered to express PSMA (HEK293-PSMA) and HEK293 cells engineered to express STEAP1 (HEK293-STEAP1) cultured in DMEM media supplemented with 10% heat-inactivated FBS at mid-log phase were counted (viability, >90%), and 100 μl containing 1500 to 2500 cells were plated into each well of a white 96-well plate (Corning, 3610). After 24 hour incubation at 37° C. in a humidified incubator with 5% CO2, two different dilutions (1:100, 1:1000) of HTAP-generated small scale hybridoma supernatants were incubated with goat anti-human IgG1-MMAF, DAR8 (final concentration 2 μg/mL) conjugated secondary antibody for 20 minutes. Samples were added to each well and incubated for 5 days. Cell viability was assessed with the CellTiter-Glo© Luminescent Cell Viability Assay (Promega, G7570) according to the manufacturer's instructions, with the exception that the plates were shaken for 15 minutes prior to reading. Luminescence was detected on a Perkin Elmer Victor X3 instrument.
Flow cytometry binding assay was also used to characterize the HTAP DVD-Igs. Specifically, HEK293-PSMA or HEK293-STEAP1 cells grown in T-75 flask (Corning, 4430720) were harvested using 5-mL enzyme-free Cell Dissociation Buffer (Invitrogen, 13151-014), washed once with 5 mL FACs buffer (BD Pharmingen, 554657), and then resuspended at 2.5×106 cells/mL in FACS buffer. A volume of 100 μL of cells, equivalent to 2.5×105 cells/well, was then dispensed into a V-bottom 96-well plate. This was followed by the addition of 100 μL of various concentrations of DVD-Ig, and the samples were incubated at 4° C. for 1 hour. Wells were washed twice with FACs buffer and then resuspended in 100 μL of 1:100 anti-human IgG antibody (PE, Southern Biotech, 2040-09) along with Far Red Live/Dead Stain at 1 μL/mL (Invitrogen, L34974A) diluted in FACs buffer. The plate was incubated at 4° C. for another 1 hour and then washed twice with FACs buffer. Cells were then resuspended in 100 μL of D-PBS (Sigma, D8537)/1% formaldehyde and analyzed on a BD FACS Canto II flow cytometer. The data collected from the cytometer was subsequently analyzed using the FlowJo flow cytometry analysis software.
20 DVD-Igs passed initial production and binding screens. Table 3 provides a summary of in vitro potency, flow cytometry binding of DVD-Igs to LNCaP.FGC, and HEK293 cells engineered to express PSMA and STEAP1.
Eight PSMA-STEAP1 DVD-Igs (Clone 2, Clone 4, Clone 9, Clone 11, Clone 12, Clone 13, Clone 14, and Clone 15) were conjugated with monomethyl auristatin E (MMAE) and evaluated in LNCaP.FGC, HEK293-STEAP1, and HEK293-PSMA cells in vitro. An anti-STEAP1-MMAE ADC and two comparable anti-PSMA-MMAE ADCs were used as controls.
Cells (LNCaP.FGC, HEK293-PSMA, and HEK293-STEAP1) were counted (viability, >90%), and 100 μL of medium containing 1500 to 2500 cells were plated into each well of a white 96-well plate (Corning, 3610). After 24 hours incubation at 37TC in a humidified incubator with 5% CO2, 50 μL of 3× stock solution of conjugate was added per well, and cells were incubated for additional 7 days. Samples were analyzed in triplicate. Cell viability was assessed with the CellTiter-Glo® Luminescent Cell Viability Assay (Promega, G7570) according to the manufacturer's instructions, with the exception that the plates were shaken for 15 minutes prior to reading. Luminescence was detected on a Perkin Elmer Victor X3 instrument. Untreated control wells were included on each plate. Cells treated with AB095 DVD-Ig, an anti-tetanus toxoid DVD-Ig, conjugated with MMAE was also included as a control.
All eight PSMA-STEAP1 DVD-Igs conjugated with MMAE showed cytotoxic activity in LNCaP.FGC cells (
In vitro cytotoxic activity of PSMA-STEAP1 DVD-Ig clone 15 conjugated with MMAE (“DVD-MMAE”) was confirmed and shown in
Seven of the eight MMAE-conjugated PSMA-STEAP1 DVD-Igs (Clone 4, Clone 9, Clone 11, Clone 12, Clone 13, Clone 14, and Clone 15) that showed in vitro cytotoxic activity were further evaluated in in vivo studies. MMAE-conjugated PSMA-STEAP1 DVD-Ig Clone 2 was not included in the in vivo studies due to it having a high percentage of aggregation.
Male NSG mice were obtained from Jackson Laboratories (Bar Harbor, ME) for both LNCaP.FGC and 22Rv1 xenograft studies. Mice were housed by treatment group at seven to eight mice per cage. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a 12-hour light:12-hour dark schedule (lights on at 06:00 hours).
To generate xenografts from LNCaP.FGC (ATCC) and from 22Rv1 (ATCC) cell lines, 5×106 viable LNCaP.FGC and 22Rv1 cells were inoculated subcutaneously into the right flank of male NSG mice, respectively. The injection volume was 0.1 mL and composed of a 1:1 mixture with Matrigel (BD, Franklin Lakes, NJ). Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the tumor volume was calculated according to the following equation: V=L×W2/2.
Mice were assigned to treatment groups so that the mean tumor volumes within groups were approximately 175-250 mm3. An anti-PSMA-MMAE ADC at Dose-1 1×, an anti-STEAP1-MMAE ADC at Dose-1 1×, and various of the PSMA/STEAP-1 DVD-Igs conjugated with MMAE described above at Dose-1 1.33×(Clone 13 was administered at Dose-1 0.87×due to limited compound availability) were administered within 24-48 hours of assignment to treatment groups based on tumor volumes. Tumor volumes were estimated two times weekly after dosing, with study groups body weights taken at the time of tumor measurements. Mice were euthanized (1) when tumor volume reached 2,000 mm3, (2) when general efficacy parameters were confidently determined, or (3) when necessitated by other animal health issues.
Parameters of Efficacy were determined as follows:
TGI (tumor growth inhibition)=1−(mean tumor volume of treatment group/mean tumor volume of treatment control group)×100 based on measurements on day of vehicle endpoint. P values are derived from Student's T test comparison of treatment group vs. vehicle control group.
TGD (tumor growth delay)=(T−C)/C×100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of treatment control group. P values are derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group.
The parameters of efficacy for all 7 PSMA-STEAP1 DVD-Ig clones conjugated with MMAE tested in the LNCaP.FGC xenograft studies are shown in Table 5B below.
PSMA-STEAP1 DVD-Ig clone 15 conjugated with MMAE (“DVD-MMAE”) showed comparable or superior activity relative to monospecific ADCs (“PSMA-MMAE” and “STEAP1-MMAE”) in 22Rv1 (
Based on the results of the in vitro screening and the in vivo study, clone 15 was selected for further evaluation and engineering. It demonstrated good potency and acceptable drug-like properties, but manifested a deamidation motif (20%) in the PSMA binding domain.
Rational design was performed to identify and remove high risk sequence liabilities. DVDs with mutations were produced and evaluated in a Biacore assay, where the kinetics of binding to PSMA resulted in the identification NT as a non-disruptive mutation away from “NG”. NT was used in this position of HCDR2 in further versions of the DVD-Ig since no significant loss of binding occurred. In order to facilitate DAR2 or DAR6 conjugation, the variable regions were cloned onto a light chain containing a Cysteine to Alanine mutation. This creates an unpaired cysteine at position C220 of the HC for conjugation of the linker drug in a facile, site-specific manner.
DVD-Ig1 was generated by performing deamidation of VH2-CDR2 of clone 15, and the sequence is shown below in Table 1.
EVQLVESGGGVVQPGRSLRLSCVAS
GFPENNYWMT
WIRQ
APGKGLEWIA
SITITGGTTYYPGSVKG
RFTISRDNSKSTL
YLQMNSLRAEDTAVYYCTR
ERYSTEYYPYYWYFDF
WGQGT
MVTVSS
GGGGSGGGGS
EVQLVQSGAEVKKPGSSVKVSCKA
SGYTFTDYYMNWVKQAPGKGLEWIG
LINPSNTGTNYNQK
FRD
KATLTVDKSTSTAYMELSSLRSEDTAVYYCAR
RAQAT
SGDY
WGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAAL
EVQLVESGGGVVQPGRSLRLSCVAS
GFPENNYWMT
WIRQA
PGKGLEWIA
SITITGGTTYYPGSVKG
RFTISRDNSKSTLY
LQMNSLRAEDTAVYYCTR
ERYSTEYYPYYWYFDF
WGQGTM
VTVSS
GFPENNYWMT
SITITGGTTYYPGSVKG
ERYSTEYYPYYWYFDE
GGGGSGGGGS
EVQLVQSGAEVKKPGSSVKVSCKAS
GYTFTDYYMN
WVKQ
APGKGLEWIG
LINPSNTGTNYNQKFRD
KATLTVDKSTSTA
YMELSSLRSEDTAVYYCAR
RAQATSGDY
WGQGTTVTVSS
GYTFTDYYMN
LINPSNTGTNYNQKFRD
RAQATSGDY
DIQMTQSPSSLSASVGDRVTITC
RTSQDIGNYLR
WFQQKPG
KSPKLMIY
DAINLAA
GVPSRFSGSGSGTDYTLTISSLQPED
VATYYC
LQHNEYPYT
FGQGTKLEIKR
GGSGGGGS
GDIQMTQ
SPSSLSASVGDRVTITCHASQNINVWLSWYQQKPGKAPKLL
IYKASNLHTGVPSRFSGSGSGTDFTFTISSLQPEDIATYYC
QQGQTYPLT
FGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGT
DIQMTQSPSSLSASVGDRVTITCRTSQDIGNYLRWFQQKPG
KSPKLMIYDAINLAAGVPSRESGSGSGTDYTLTISSLQPED
VATYYCLQHNEYPYTFGQGTKLEIK
RTSQDIGNYLR
DAINLAA
LQHNEYPYT
GGSGGGGSG
DIQMTQSPSSLSASVGDRVTITC
HASQNINVWLS
WYQQKPG
KAPKLLIY
KASNLHT
GVPSRFSGSGSGTDFTFTISSLQPED
IATYYC
QQGQTYPLT
FGGGTKVEIK
HASQNINVWLS
KASNLHT
QQGQTYPLT
EVQLVESGGGVVQPGRSLRLSCVAS
GFPENNYWMT
WIRQ
APGKGLEWIA
SITITGGTTYYPGSVKG
RFTISRDNSKSTL
YLQMNSLRAEDTAVYYCTR
ERYSTEYYPYYWYFDF
WGQGT
MVTVSSGGGGSGGGGSEVQLVQSGAEVKKPGSSVKVSCKA
SGYTFTDYYMN
WVKQAPGKGLEWIG
LINPSNTGTNYNQK
FRD
KATLTVDKSTSTAYMELSSLRSEDTAVYYCAR
RAQAT
SGDY
WGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAAL
As shown in the examples above, DVD-Ig1 was identified based on expression, drug-like properties, in vitro dual binding potency, in vivo activity, and absence of sequence liability. It was then used to generate a Top1i drug conjugate.
The structure of Top1i drug-linker is shown as follows:
509.9 mL antibody solution (11.34 mg/mL) in PBS was added to 2.0 mL of 0.5 M pH 7.5 EDTA solution. A solution of diphenyl-phosphino-4-benzoic acid (10 mM, 18.38 mL, 2.5 eq) in DMA was then added and the mixture was kept at 4° C. overnight (18 hours) for reduction. 51.6 mL (20% v/v) 100 mM pH 8.5 sodium bicarbonate buffer was added to the reduced antibody, and then treated with the drug-linker solution (10 mM, 11.4 mL, 4.0 equivalence to antibody) and kept at 4° C. for 60 minutes. Crude conjugate was purified to remove excessive drug-linker and other small-molecule reagent.
Hydrophobic interaction chromatography was performed using TSK-GEL® Ether-5PW Column (TOSOH Bioscience, 2.0 mm ID×7.5 cm, 10 PM) at 30° C. ADC was 50% Buffer A, loaded and eluted with 10% B for 1 minute, followed by a gradient of 10% B to 100% B in 30 minutes. Mobile Phase A was 25 mM sodium phosphate buffer, 1.5 M ammonium sulfate, pH 7.0; Mobile Phase B was mixture of 75% 25 mM sodium phosphate buffer pH 7.0 and 25% isopropanol, and flow rate was 0.1 mL/min. Data is shown in
Size exclusion chromatography was performed using Waters ACQUITY UPLC® Protein BEH SEC Column (200 Å, 1.7 μm, 4.6 mm×150 mm) with 100 mM sodium phosphate, 150 mM sodium chloride, 10% volume/volume acetonitrile, pH 7.0 at a flow rate of 0.4 mL/minute. The peak area absorbance at 280 nm was determined for each of the high molecular weight and monomeric eluents by integration of the area under the curve. The % aggregate fraction of the conjugate sample was determined by dividing the peak area absorbance at 280 nm for the high molecular weight eluent by the sum of the peak area absorbances at 280 mM of the high molecular weight and monomeric eluents multiplied by 100%. Percent aggregation was determined at 2.7% (
LC-MS analysis was performed using an Agilent 1100 HPLC system interfaced to an Agilent LC/MSD TOF 6220 ESI mass spectrometer. The ADC was reduced with 25 mM (final concentration) Bond-Breaker® TCEP solution (Thermo Scientific, Rockford, IL), loaded onto a Protein Microtrap (Michrom Bioresorces, Auburn, CA) desalting cartridge, and eluted with a gradient of 10% B to 75% B in 0.2 minutes at ambient temperature. Mobile phase A was H2O with 0.1% formic acid, mobile phase B acetonitrile with 0.1% formic acid, and flow rate was 0.2 mL/minute. Electrospray-ionization time-of-flight mass spectra of the co-eluting light and heavy chains were acquired using Agilent MassHunter™ acquisition software. The extracted intensity vs. m/z spectrum was deconvoluted using the Maximum Entropy feature of MassHunterp software to determine the mass of each reduced antibody fragment. DAR was calculated from the deconvoluted spectrum by summing intensities of the naked and modified peaks for the light chain and heavy chain, normalized by multiplying intensity by the number of drugs attached. The summed, normalized intensities were divided by the sum of the intensities, and the summing results for two light chains and two heavy chains produced a final average DAR value for the full ADC.
MS data:
Internalization and in vitro cytotoxicity of DVD-Ig1 drug conjugate was characterized.
Cells were maintained in a humidified 37° C. incubator with 5% CO2. Exponentially growing LNCaP.FGC cells were detached using 0.25% trypsin-EDTA (Sigma, T4049) and 20,000 cells/well were plated in 96 well-plates (Corning, 3610) in 50 μl cell culture media. Next day, antibody conjugates (PSMA-STEAP1-Top1i, PSMA-Top1i, and STEAP1-Top1i) and FabFluor reagent (Sartorius, 4722) were mixed at a molar ratio of 1:3 in media, for 15 minutes. The labeled antibodies were serially diluted and added to each well in triplicates. The plates were placed in Incucyte Zoom (Sartorius) for image collection for up to 24 hours. Collected images were processed and analyzed. The final data was exported to the GraphPad Prism software for graph generation.
The intracellular accumulation assay was used to measure mean intracellular concentrations of unconjugated Top1i. Specifically, 2 million LNCaP.FGC cells in 40 ml medium were plated in a T150 flask 24 hours prior to the treatment. Each T150 flask was treated with 0 nM, 1 nM, 10 nM, or 30 nM of PSMA-STEAP1-Top1i, PSMA-Top1i, or STEAP1-Top1i. Samples were collected at timepoints 4, 6, and 24 hours. Two technical replicates at each time point were used. Cells without treatment (0 nM) was the control. At each time point, cells were washed twice with PBS and detached with trypsin to prepare single cell suspension. Live cell count and cell diameter were determined from the single cell suspension to calculate total cellular volume (in liter). LC-MS analysis was conducted to determine the amount of intracellular small molecule (in nanomoles). The LC-MS method quantified total cell associated molecules which were not covalently bound to any entity, i.e., sum of bound and unbound molecules in the cell. All molecules covalently bound to cellular proteins/lipids were not extracted and measured. Top1i compound has two forms: the active closed ring (Lactone) form at low pH, or inactive open ring (Carboxylate) form. Protein was precipitated with acidic dilution when preparing samples for LC-MS analysis, because lower pH pushed equilibrium to active closed ring form. During assay, media pH became acidic. The LC-MS bioanalysis converted majority of unconjugated payload to the closed ring status, and hence measured “total payload” (i.e., open form and closed form) irrespective of their original forms present in the sample.
Mean intracellular concentration (in nanomolar) was then calculated based on the total cellular volume and the amount of intracellular small molecule. Additional calculation details are shown below:
LNCaP.FGC cells were dissociated using 0.25% trypsin-EDTA (Sigma, T4049) and counted (viability >90%). 100 mL of growth medium containing 2500 cells were plated into each well of a white 96-well plate (Corning, 3610). After 24-hour incubation at 37° C. in a humidified incubator with 5% CO2, 50 mL of 3× stock solution of conjugate or payload were added per well, and cells were incubated for an additional 7 days. Samples were analyzed in triplicate. Cell viability was assessed with the CellTiter-Glo Luminescent Cell Viability Assay (Promega, G7570) according to the manufacturer's instructions, with the exception that the plates were shaken for 15 minutes prior to reading. Luminescence was detected on a Perkin Elmer Victor X3 instrument. Untreated control wells were included on each plate. Percent of control was calculated using the following formula: 100×(treated)/(control). Half-maximal inhibition (IC50) values were calculated using GraphPad Prism software.
PSMA-STEAP1-Top1i also showed superior anti-proliferative activity than either monospecific ADC, PSMA-Top1i or STEAP-Top1i (
DVD-Ig1 conjugated with Top1i (“PSMA-STEAP1-Top1i”) showed efficacy in prostate tumors that express STEAP1 and PSMA in xenograft models in vivo.
Male NSG mice were obtained from Jackson Laboratories (Bar Harbor, ME) for the LNCaP.FGC xenograft study. Male CB17.SCID (Charles River Laboratories, Wilmington, MA) mice were used for the 22Rv1 xenograft study. Mice were housed by treatment group at seven to eight mice per cage. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a 12-hour light:12-hour dark schedule (lights on at 06:00 hours).
To generate xenografts from LNCaP.FGC (ATCC) and from 22Rv1 (ATCC) cell lines, 5×106 viable LNCaP.FGC and 22Rv1 cells were inoculated subcutaneously into the right flank of male NSG mice and male CB17.SCID mice, respectively. The injection volume was 0.1 mL and composed of a 1:1 mixture with Matrigel (BD, Franklin Lakes, NJ). Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the tumor volume was calculated according to the following equation: V=L×W2/2.
Mice were assigned to treatment groups so that the mean tumor volumes within groups were approximately 175-250 mm3. For the LNCaP.FGC xenograft study, PSMA-Top1i at Dose-2 (1×), STEAP1-Top1i at Dose-2 (1×), and PSMA-STEAP1-Top1i at Dose-2 (1.3×) were administered within 24-48 hours of reassignment. For the LNCaP.FGC xenograft study, PSMA-Top1i at Dose-3 (1×), STEAP1-Top1i at Dose-3 (1×), and PSMA-STEAP1-Top1i at Dose-3 (1.3×) were administered within 24-48 hours of reassignment. Tumor volumes were estimated two times weekly after dosing, with study groups body weights taken at the time of tumor measurements. Mice were euthanized (1) when tumor volume reached 2,000 mm3, (2) when general efficacy parameters were confidently determined or (3) when other animal health issues were observed.
PSMA-STEAP1-Top1i was highly efficacious against LNCaP.FGC subcutaneous flank tumors when administered as a single dose at Dose-2 (1.3×) (
For 22Rv1, a CDX model which also expressed high levels of both PSMA and STEAP1 based on RNA expression, PSMA-STEAP1-Top1i was also highly efficacious at Dose-3 (1.3×) (
These data indicate that DVD-Ig1 disclosed herein has activity comparable or better than that of individual ADCs, and that a single dose of PSMA-STEAP1-Top1i supports sustained tumor regression.
Embodiment 1. A dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
Embodiment 2. The DVD-Ig drug conjugate of embodiment 1, wherein the first heavy chain variable domain (VH1) comprises the amino acid sequence of SEQ ID NO: 2; the second heavy chain variable domain (VH2) comprises the amino acid sequence of SEQ ID NO: 7; the first light chain variable domain (VL1) comprises the amino acid sequence of SEQ ID NO: 12; and the second light chain variable domain (VL2) comprises the amino acid sequence of SEQ ID NO: 17.
Embodiment 3. The DVD-Ig drug conjugate of embodiment 1 or 2, wherein the first peptide linker (L1) comprises the amino acid sequence GGGGSGGGGS (SEQ ID NO: 6).
Embodiment 4. The DVD-Ig drug conjugate of any one of embodiments 1-3, wherein the second peptide linker (L2) comprises the amino acid sequence GGSGGGGSG (SEQ ID NO: 16).
Embodiment 5. The DVD-Ig drug conjugate of any one of embodiments 1-4, wherein the IgG1 heavy chain constant region (CH1CH2CH3) comprises the amino acid sequence of SEQ ID NO: 21 or 23.
Embodiment 6. The DVD-Ig drug conjugate of any one of embodiments 1-5, wherein the kappa light chain constant region (Cκ) comprises the amino acid sequence of SEQ ID NO: 22.
Embodiment 7. The DVD-Ig drug conjugate of any one of embodiments 1-6, wherein each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24; and wherein each light chain comprises the amino acid sequence of SEQ ID NO: 11.
Embodiment 8. The DVD-Ig drug conjugate of any one of embodiments 1-7, wherein the first heavy chain and the second heavy chain are connected by two disulfide bridges, and wherein the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering.
Embodiment 9. The DVD-Ig drug conjugate of any one of embodiments 1-8, wherein the Top1i linker-Drugs are conjugated to the cysteines at position C220 of the first and the second heavy chains of the DVD-Ig according to EU numbering.
Embodiment 10. The DVD-Ig drug conjugate of any one of embodiments 1-9, wherein n is 1.
Embodiment 11. The DVD-Ig drug conjugate of any one of embodiments 1-9, wherein n is 2.
Embodiment 12. A dual-variable domain immunoglobulin (DVD-Ig) drug conjugate comprising the following structure:
Embodiment 13. A DVD-Ig that binds human STEAP1 and human PSMA, comprising two identical heavy chains and two identical light chains,
Embodiment 14. The DVD-Ig of embodiment 13, wherein the first heavy chain variable domain (VH1) comprises the amino acid sequence of SEQ ID NO: 2; the second heavy chain variable domain (VH2) comprises the amino acid sequence of SEQ ID NO: 7; the first light chain variable domain (VL1) comprises the amino acid sequence of SEQ ID NO: 12; and the second light chain variable domain (VL2) comprises the amino acid sequence of SEQ ID NO: 17.
Embodiment 15. The DVD-Ig of embodiment 13 or 14, wherein the first peptide linker (L1) comprises the amino acid sequence GGGGSGGGGS (SEQ ID NO: 6).
Embodiment 16. The DVD-Ig of any one of embodiments 13-15, wherein the second peptide linker (L2) comprises the amino acid sequence GGSGGGGSG (SEQ ID NO: 16).
Embodiment 17. The DVD-Ig of any one of embodiments 13-16, wherein the IgG1 heavy chain constant region (CH1CH2CH3) comprises the amino acid sequence of SEQ ID NO: 21 or 23.
Embodiment 18. The DVD-Ig of any one of embodiments 13-17, wherein the kappa light chain constant region (Cκ) comprises the amino acid sequence of SEQ ID NO: 22.
Embodiment 19. The DVD-Ig of any one of embodiments 13-18, wherein each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24; and wherein each light chain comprises the amino acid sequence of SEQ ID NO: 11.
Embodiment 20. The DVD-Ig of any one of embodiments 13-19, wherein the first heavy chain and the second heavy chain are connected by two disulfide bridges, and wherein the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering.
Embodiment 21. A DVD-Ig that binds human STEAP1 and human PSMA comprising two identical heavy chains and two identical light chains, wherein each of the heavy chains comprise the amino acid sequence of SEQ ID NO: 1 or 24; and wherein each light chain comprises the amino acid sequence of SEQ ID NO: 11; and wherein the first heavy chain and the second heavy chain are connected by two disulfide bridges, and wherein the two disulfide bridges are between the cysteine in position 226 of the first heavy chain and the cysteine in position 226 of the second heavy chain, and between the cysteine in position 229 of the first heavy chain and the cysteine in position 229 of the second heavy chain according to EU numbering.
Embodiment 22. A nucleic acid encoding a heavy chain comprising an amino acid sequence of SEQ ID NO: 1 or 24, and/or a light chain comprising an amino acid sequence of SEQ ID NO: 11.
Embodiment 23. A vector comprising a nucleic acid of embodiment 22.
Embodiment 24. A host cell comprising a nucleic acid of embodiment 22, or a vector of embodiment 23.
All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.
While various specific embodiments have been illustrated and described, it will be appreciated that various changes can be made without departing from the spirit and scope of the invention(s).
This application claims the benefit of U.S. Provisional Application No. 63/604,884, filed Nov. 30, 2023, and U.S. Provisional Application No. 63/550,451, filed Feb. 6, 2024, the contents of which are hereby incorporated by reference in their entirety.
Number | Date | Country | |
---|---|---|---|
63550451 | Feb 2024 | US | |
63604884 | Nov 2023 | US |