WO2023245078A1 - Anticorps anti-parvovirus et leurs utilisations - Google Patents

Anticorps anti-parvovirus et leurs utilisations Download PDF

Info

Publication number
WO2023245078A1
WO2023245078A1 PCT/US2023/068460 US2023068460W WO2023245078A1 WO 2023245078 A1 WO2023245078 A1 WO 2023245078A1 US 2023068460 W US2023068460 W US 2023068460W WO 2023245078 A1 WO2023245078 A1 WO 2023245078A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
antibody
binding fragment
amino acid
parvovirus
Prior art date
Application number
PCT/US2023/068460
Other languages
English (en)
Inventor
Davide Corti
Barbara GUARINO
Original Assignee
Humabs Biomed Sa
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 Humabs Biomed Sa filed Critical Humabs Biomed Sa
Publication of WO2023245078A1 publication Critical patent/WO2023245078A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/081Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • parvovirus B19 is one of the only members of the Parvoviridae family infecting humans.
  • the B 19 virus is also associated with more severe diseases, including hydrops fetalis and fetal death, aplastic crises, polyarthralgia and arthritis and pure red-cell aplasia.
  • the current treatment for patients suffering from persistent B 19 virus infection is administration of human immunoglobulin preparations, e.g., intravenous immunoglobulin therapy (IVIG), which have limited efficacy and toxicity risks.
  • IVIG intravenous immunoglobulin therapy
  • Human immunoglobulins are also scarce, as it is challenging to find human donors, and costly.
  • the use of non-human, animal immunoglobulins (e.g., equine) is affected by challenges such as a limited production, animal welfare concerns, and safety issues.
  • Figure 1 shows results of stimulated B cells from healthy donor tonsils screened for binding to VPl/VP2-expressing VLPs. 43% of the tonsil donors scored positive and 3 were selected for isolation of monoclonal antibodies (mAbs) (outlined).
  • Figure 2A-2D show results of an immunodominance study in antigen-specific B-cell memory repertoire analysis (AMBRA) supernatants and different collections of sera.
  • Figure 2A shows frequency of positivity from AMBRA supernatants on VP1/VP2 and VP2 virus-like particles (VLPs).
  • Figures 2B-2C show endpoint VP 1 vs VP2 from 60 sera collected in Bologna, Italy ( Figure 2B) or 111 sera collected in Bellinzona, Switzerland ( Figure 2C).
  • Antibody values were expressed as endpoint titers defined as the highest dilution at which the OD was higher than twice the background OD.
  • Figure 2D shows inhibition of competition in sera collected in Bellinzona with PAB5 (VP1) biotinylated vs PAB18 (VP2) biotinylated.
  • Figures 3A-3D show characterization of selected mAbs.
  • Figure 3A provides a table showing parvovirus neutralization activity of mAb, the light chain isotype of as mAb determined by enzyme-linked immunosorbent assay (ELISA), the germline usage for V-D-J (VH) and V-J (VL) genes defined using the ImMunoGeneTics information system (IMGT) database and result of Western blot binding to reduced and non-reduced antigen.
  • Figures 3B-3D show binding of various mAbs isolated from immortalized IgG+ memory B cells as assessed by ELISA.
  • Figure 3C shows mAbs binding to VP1/VP2 and VP2 VLPs (z.e., VP2- specific) (PAC57, PAC58, and PAC69 are not recombinant).
  • Figure 3D shows IgG and Fab of PAB5 mAb binding to VP1/VP2 VLPs.
  • Figures 4A-4C show results from comparison of binding and blockade of binding studies (BOB).
  • Figure 4A shows binding of IVIG, PAB5 and PAB18 mAbs to VP1/VP2 VLPs by ELISA.
  • Figures 5A-5E show results from cross-competition studies and epitope mapping.
  • Figure 5A shows cross-competition studies of anti-VPl mAbs against biotinylated anti-VPl mAbs.
  • Figure 5B shows cross-competition studies of anti-VP2 mAbs against biotinylated anti-VP2 mAbs.
  • Figure 5C shows an intensity plot with averaged spot intensities of PAB5-rIgGl (lOpg/ml) on a conformational peptide microarray containing 696 different cyclic constrained peptides.
  • Figure 5D shows an intensity plot with averaged spot intensities of PAA19-rIgGl (500 pg/ml) on a conformational peptide microarray containing 1,677 different cyclic constrained peptides.
  • the intensity plots were correlated with peptide and intensity maps as well as with visual inspection of the microarray scans to identify the epitopes of the human mAbs. In case it was not clear if a certain amino acid contributed to antibody binding, the corresponding letter was written in gray (lighter font).
  • Figure 5E shows an amino acid plot of PAB5-rIgGl against the microarray containing conformational peptides with 119 variants of the wild type peptide: the plot was calculated by dividing the spot intensity of a given peptide (e.g., 1 YPYDVQDYA 9 (SEQ ID NO.:44)) by the spot intensity of the native epitope ( 1 YPYDVPDYA 9 (SEQ ID NO.:45)). The position of an amino acid at a given position, thus, reflected the intensity ratio compared to the wild type amino acid at the same position.
  • a given peptide e.g., 1 YPYDVQDYA 9 (SEQ ID NO.:44)
  • 1 YPYDVPDYA 9 SEQ ID NO.:45
  • Figures 6A-6C show data on neutralization of B 19 by selected VP1 and VP2 mAbs.
  • Figure 6A shows fluorescence-activated cell sorting (FACS) data on EPC primary cells.
  • Figure 6B shows percentage neutralization by the selected VP1 and VP2 mAbs.
  • Figure 6C shows qPCR data on EPC primary cells.
  • “Grifols” anti-parvovirus immune globulin preparation (for intravenous administration (IVIG)) from Grifols Pharmaceutical.
  • Figure 7 shows modeled structures of VP1 receptor-binding domain (RBD) having different mutations.
  • Figures 8A-8C show amino acid sequences and certain polynucleotide sequences for variable domains of anti-parvovirus antibodies PAB18 (8A), PAB5 (8B), and PAA19 (8C).
  • Figures 9A-9C show neutralization of infection by antibodies and by an antibody combination, as described in Example 6.
  • Figures 9A and 9B show results from different concentrations of antibodies.
  • Figure 9C shows results from antibodies, antibody Fabs, or combinations, at lug/ml of antibody, Fab, or combination.
  • Figures 10A-10B show neutralization of infection (IC80, ug/ml shown as foldchange vs. IVIG) by antibodies PAB5, PAA19, and PAB18, and by PAB5 Fab, as described in Example 6.
  • “Grifols” anti-parvovirus immune globulin preparation (for intravenous administration (IVIG)) from Grifols Pharmaceutical.
  • Figure 11 shows titers of VPl/VP2-specific and VP2-specific antibodies in sera from human patients with chronic parvovirus B19 infection (left) and from healthy human donors (right). Endpoint VP 1 vs VP2 from antibody values were expressed as endpoint titers defined. as the highest dilution at which the OD was higher than twice the background OD. These data illustrate that most anti -parvovirus antibodies in sera are directed to B19 VP2.
  • Figure 12 shows results from AMBRA analysis of human tonsil samples, illustrating that antibody response against B 19 VP1 is rare.
  • the bottom graph shows frequency of positivity from AMBRA supernatants on VP1 and VP2 (tonsils T 1-2- 12- 21-24-26-28-32-36).
  • antibodies and antigen-binding fragments that can bind to and, in certain embodiments, can potently neutralize infection by parvovirus, such as parvovirus Bl 9.
  • a fragment antigen-binding (Fab) of an antibody or antigen-binding fragment is sufficient for potent neutralization of infection by a parvovirus, such as parvovirus Bl 9.
  • polynucleotides that encode the antibodies and antigen-binding fragments, vectors, host cells, and related compositions, as well as methods of using the antibodies, polynucleotides, vectors, host cells, and related compositions to treat (e.g., reduce, delay, eliminate, or prevent) and/or to diagnose a parvovirus infection in a subject and/or in the manufacture of a medicament for treating a parvovirus infection in a subject.
  • Non-limiting examples of anti-parvovirus antibodies of the present disclosure include the human antibodies “PAB5”, PAA19”, and “PAB18”.
  • PAB5 is a potent neutralizing antibody that binds in a conserved region of B19 uVPl.
  • PAA19 and PAB18 are also neutralizing antibodies and bind to distinct antigenic sides in VP2.
  • Some embodiments provide an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB5. Some embodiments provide an antibody or antigenbinding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAA19. Some embodiments provide an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB18. Also provided are antibodies and antigen-binding fragments comprising one or more CDR, one or more variant CDR, and/or one or more other variations in a VH and/or in a VL, relative to PAB5, PAA19, or PAB18.
  • composition or combination that comprises two or more different presently disclosed antibodies or antigen-binding fragments.
  • some embodiments provide a composition or combination comprising: (i) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB5, and (ii) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAA19.
  • compositions or combination comprising: (i) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB5, and (ii) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB18.
  • compositions or combination comprising: (i) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAB18, and (ii) an antibody or antigen-binding fragment comprising the six CDRs, and optionally comprising the VH and VL amino acid sequences, of antibody PAA19.
  • an antibody or antigen-binding fragment comprises a heavy chain and the heavy chain comprises one or more mutations for extending in vivo half-life (e.g., in a human) of the antibody or antigen-binding fragment.
  • a heavy chain comprises the mutations M428L and N434S (EU numbering).
  • any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness are to be understood to include any integer within the recited range, unless otherwise indicated.
  • the term “about” means ⁇ 20% of the indicated range, value, or structure, unless otherwise indicated. It should be understood that the terms “a” and “an” as used herein refer to “one or more" of the enumerated components.
  • a protein domain, region, or module e.g., a binding domain
  • a protein "consists essentially of' a particular amino acid sequence when the amino acid sequence of a domain, region, module, or protein includes extensions, deletions, mutations, or a combination thereof (e.g., amino acids at the amino- or carboxy -terminus or between domains) that, in combination, contribute to at most 20% (e.g., at most 15%, at most 10%, at most 8%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2% or at most 1%) of the length of a domain, region, module, or protein and do not substantially affect (i.e., do not reduce the activity by more than 50%, such as no more than 40%, no more than 30%, no more than 25%, no more than 20%, no more than 15%, no more than 10%, no more than 5%, or no more than 1%) the activity of the domain(s), region(s), module(s), or protein (e.g., the
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, y-carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refer to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a-carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refer to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • mutation refers to a change in the sequence of a nucleic acid molecule or polypeptide molecule as compared to a reference or wild-type nucleic acid molecule or polypeptide molecule, respectively.
  • a mutation can result in several different types of change in sequence, including substitution, insertion or deletion of nucleotide(s) or amino acid(s).
  • a “conservative substitution” refers to amino acid substitutions that do not significantly affect or alter binding characteristics of a particular protein. Generally, conservative substitutions are ones in which a substituted amino acid residue is replaced with an amino acid residue having a similar side chain. Conservative substitutions include a substitution found in one of the following groups: Group 1 : Alanine (Ala or A), Glycine (Gly or G), Serine (Ser or S), Threonine (Thr or T); Group 2: Aspartic acid (Asp or D), Glutamic acid (Glu or Z); Group 3 : Asparagine (Asn or N), Glutamine (Gin or Q); Group 4: Arginine (Arg or R), Lysine (Lys or K), Histidine (His or H); Group 5: Isoleucine (He or I), Leucine (Leu or L), Methionine (Met or M), Valine (Vai or V); and Group 6: Phenylalanine (Phe or F), Tyrosine (Tyr
  • amino acids can be grouped into conservative substitution groups by similar function, chemical structure, or composition (e.g., acidic, basic, aliphatic, aromatic, or sulfur-containing).
  • an aliphatic grouping may include, for purposes of substitution, Gly, Ala, Vai, Leu, and He.
  • Other conservative substitutions groups include: sulfur-containing: Met and Cysteine (Cys or C); acidic: Asp, Glu, Asn, and Gin; small aliphatic, nonpolar or slightly polar residues: Ala, Ser, Thr, Pro, and Gly; polar, negatively charged residues and their amides: Asp, Asn, Glu, and Gin; polar, positively charged residues: His, Arg, and Lys; large aliphatic, nonpolar residues: Met, Leu, He, Vai, and Cys; and large aromatic residues: Phe, Tyr, and Trp. Additional information can be found in Creighton (1984) Proteins, W.H. Freeman and Company.
  • protein or “polypeptide” refers to a polymer of amino acid residues. Proteins apply to naturally occurring amino acid polymers, as well as to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, and non-naturally occurring amino acid polymers. Variants of proteins, peptides, and polypeptides of this disclosure are also contemplated.
  • variant proteins, peptides, and polypeptides comprise or consist of an amino acid sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.9% identical to an amino acid sequence of a defined or reference amino acid sequence as described herein.
  • a variant polypeptide e.g.
  • antibody or antigen-binding fragment comprises one or more substitutions, one or more of which is optionally a conservative substitution and/or is a substitution to a germline-encoded amino acid (see Figure 3 A for VH germline gene usage of certain antibodies fo the present disclosure).
  • Any polypeptide of this disclosure can, as encoded by a polynucleotide sequence, comprise a “signal peptide” (also known as a leader sequence, leader peptide, or transit peptide).
  • Signal peptides target newly synthesized polypeptides to their appropriate location inside or outside the cell.
  • a signal peptide may be removed in whole or in part from the polypeptide during or once localization or secretion is completed.
  • polypeptides that have a(n, e.g., full-length) signal peptide can be referred to as a “pre-protein” and polypeptides having their signal peptide removed - at least in part - can be referred to as “mature” proteins or polypeptides.
  • an antibody or antigenbinding fragment is a mature protein or a pre-protein.
  • Nucleic acid molecule or “polynucleotide” or “polynucleic acid” refers to a polymeric compound including covalently linked nucleotides, which can be made up of natural subunits (e.g., purine or pyrimidine bases) or non-natural subunits (e.g., morpholine ring).
  • Purine bases include adenine, guanine, hypoxanthine, and xanthine
  • pyrimidine bases include uracil, thymine, and cytosine.
  • Nucleic acid molecules include polyribonucleic acid (RNA), which includes mRNA, microRNA, siRNA, viral genomic RNA, and synthetic RNA, and polydeoxyribonucleic acid (DNA, also referred to as deoxyribonucleic acid), which includes cDNA, genomic DNA, and synthetic DNA, either of which may be single or double stranded. If single-stranded, the nucleic acid molecule may be the coding strand or non-coding (anti-sense) strand.
  • a nucleic acid molecule encoding an amino acid sequence includes all nucleotide sequences that encode the same amino acid sequence.
  • nucleotide sequences may also include intron(s) to the extent that the intron(s) would be removed through co- or post-transcriptional mechanisms.
  • different nucleotide sequences may encode the same amino acid sequence as the result of the redundancy or degeneracy of the genetic code, or by splicing.
  • the polynucleotide comprises a modified nucleoside, a cap-1 structure, a cap-2 structure, or any combination thereof.
  • the polynucleotide comprises a pseudouridine, a N6-methyladenonsine, a 5- methylcytidine, a 2-thiouridine, or any combination thereof.
  • the pseudouridine comprises N1 -methylpseudouridine.
  • Variant nucleic acid molecules are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and are preferably at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.9% identical to a nucleic acid molecule of a defined or reference polynucleotide as described herein, or that hybridize to a polynucleotide under stringent hybridization conditions of 0.015M sodium chloride, 0.0015M sodium citrate at about 65-68°C or 0.015M sodium chloride, 0.0015M sodium citrate, and 50% formamide at about 42°C. Nucleic acid molecule variants retain the capacity to encode a binding domain thereof having a functionality described herein, such as binding a target molecule.
  • Percent sequence identity refers to a relationship between two or more sequences, as determined by comparing the sequences. Preferred methods to determine sequence identity are designed to give the best match between the sequences being compared. For example, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment). Further, non-homologous sequences may be disregarded for comparison purposes. The percent sequence identity referenced herein is calculated over the length of the reference sequence, unless indicated otherwise. Methods to determine sequence identity and similarity can be found in publicly available computer programs.
  • Sequence alignments and percent identity calculations may be performed using a BLAST program e.g., BLAST 2.0, BLASTP, BLASTN, or BLASTX).
  • the mathematical algorithm used in the BLAST programs can be found in Altschul et al., Nucleic Acids Res. 25:3389-3402, 1997.
  • sequence analysis software is used for analysis, the results of the analysis are based on the "default values" of the program referenced. "Default values" mean any set of values or parameters which originally load with the software when first initialized.
  • Clustal W MAFFT, Clustal Omega
  • AlignMe Praline
  • GAP BESTFIT
  • Needle EMBOSS
  • Stretcher EMBOSS
  • GGEARCH2SEQ Water
  • EMBOSS Matcher
  • LALIGN SSEARCH2SEQ.
  • a global alignment algorithm such as a Needleman and Wunsch algorithm, can be used to align two sequences over their entire length, maximizing the number of matches and minimizes the number of gaps. Default values can be used.
  • scoring matrices can be used that assign positive scores for some non-identical amino acids (e.g., conservative amino acid substitutions, amino acids with similar physio-chemical properties, and/or amino acids that exhibit frequent substitutions in orthologs, homologs, or paralogs).
  • non-identical amino acids e.g., conservative amino acid substitutions, amino acids with similar physio-chemical properties, and/or amino acids that exhibit frequent substitutions in orthologs, homologs, or paralogs.
  • scoring matrices include PAM30, PAM70, PAM250, BLOSUM45, BLOSUM50, BLOUM62, BLOSUM80, and BLOSUM90.
  • sequence analysis software is used for analysis, the results of the analysis are based on the “default values” of the program referenced. “Default values” mean any set of values or parameters which originally load with the software when first initialized.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally occurring nucleic acid or polypeptide present in a living animal is not isolated, but the same nucleic acid or polypeptide, separated from some or all of the co-existing materials in the natural system, is isolated.
  • nucleic acid could be part of a vector and/or such nucleic acid or polypeptide could be part of a composition (e.g., a cell lysate), and still be isolated in that such vector or composition is not part of the natural environment for the nucleic acid or polypeptide.
  • isolated can, in some embodiments, also describe an antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition that is outside of a human body. In certain embodiments, an isolated antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition is provided.
  • gene means the segment of DNA or RNA involved in producing a polypeptide chain; in certain contexts, it includes regions preceding and following the coding region (e.g., 5’ untranslated region (UTR) and 3’ UTR) as well as intervening sequences (introns) between individual coding segments (exons).
  • regions preceding and following the coding region e.g., 5’ untranslated region (UTR) and 3’ UTR
  • intervening sequences introns between individual coding segments (exons).
  • a “functional variant” refers to a polypeptide or polynucleotide that is structurally similar or substantially structurally similar to a parent or reference compound of this disclosure, but differs slightly in composition (e.g., one base, atom or functional group is different, added, or removed, or one or more amino acid or nucleotide is substituted), such that the polypeptide or encoded polypeptide is capable of performing at least one function of the parent polypeptide with at least 50% efficiency, preferably at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.9%, or 100% level of activity of the parent polypeptide.
  • a functional variant of a polypeptide or encoded polypeptide of this disclosure has "similar binding,” “similar affinity” or “similar activity” when the functional variant displays no more than a 50% reduction in performance in a selected assay as compared to the parent or reference polypeptide, such as an assay for measuring binding affinity (e.g., Biacore® or tetramer staining measuring an association (Ka) or a dissociation (KD) constant).
  • binding affinity e.g., Biacore® or tetramer staining measuring an association (Ka) or a dissociation (KD) constant.
  • a “functional portion” or “functional fragment” refers to a polypeptide or polynucleotide that comprises only a domain, portion or fragment of a parent or reference compound, and the polypeptide or encoded polypeptide retains at least 50% activity associated with the domain, portion or fragment of the parent or reference compound, preferably at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.9%, or 100% level of activity of the parent polypeptide, or provides a biological benefit (e.g., effector function).
  • a biological benefit e.g., effector function
  • a “functional portion” or “functional fragment” of a polypeptide or encoded polypeptide of this disclosure has “similar binding” or “similar activity” when the functional portion or fragment displays no more than a 50% reduction in performance in a selected assay as compared to the parent or reference polypeptide (preferably no more than 20% or 10%, or no more than a log difference as compared to the parent or reference with regard to affinity).
  • the term "engineered,” “recombinant,” or “non-natural” refers to an organism, microorganism, cell, nucleic acid molecule, or vector that includes at least one genetic alteration or has been modified by introduction of an exogenous or heterologous nucleic acid molecule, wherein such alterations or modifications are introduced by genetic engineering (i.e., human intervention).
  • Genetic alterations include, for example, modifications introducing expressible nucleic acid molecules encoding functional RNA, proteins, fusion proteins or enzymes, or other nucleic acid molecule additions, deletions, substitutions, or other functional disruption of a cell’s genetic material. Additional modifications include, for example, non-coding regulatory regions in which the modifications alter expression of a polynucleotide, gene, or operon.
  • heterologous or non-endogenous or exogenous refers to any gene, protein, compound, nucleic acid molecule, or activity that is not native to a host cell or a subject, or any gene, protein, compound, nucleic acid molecule, or activity native to a host cell or a subject that has been altered.
  • Heterologous, non-endogenous, or exogenous includes genes, proteins, compounds, or nucleic acid molecules that have been mutated or otherwise altered such that the structure, activity, or both is different as between the native and altered genes, proteins, compounds, or nucleic acid molecules.
  • heterologous, non-endogenous, or exogenous genes, proteins, or nucleic acid molecules may not be endogenous to a host cell or a subject, but instead nucleic acids encoding such genes, proteins, or nucleic acid molecules may have been added to a host cell by conjugation, transformation, transfection, electroporation, or the like, wherein the added nucleic acid molecule may integrate into a host cell genome or can exist as extra-chromosomal genetic material (e.g., as a plasmid or other self-replicating vector).
  • homologous or homolog refers to a gene, protein, compound, nucleic acid molecule, or activity found in or derived from a host cell, species, or strain.
  • a heterologous or exogenous polynucleotide or gene encoding a polypeptide may be homologous to a native polynucleotide or gene and encode a homologous polypeptide or activity, but the polynucleotide or polypeptide may have an altered structure, sequence, expression level, or any combination thereof.
  • a non-endogenous polynucleotide or gene, as well as the encoded polypeptide or activity may be from the same species, a different species, or a combination thereof.
  • a nucleic acid molecule or portion thereof native to a host cell will be considered heterologous to the host cell if it has been altered or mutated, or a nucleic acid molecule native to a host cell may be considered heterologous if it has been altered with a heterologous expression control sequence or has been altered with an endogenous expression control sequence not normally associated with the nucleic acid molecule native to a host cell.
  • heterologous can refer to a biological activity that is different, altered, or not endogenous to a host cell.
  • heterologous nucleic acid molecule can be introduced into a host cell as separate nucleic acid molecules, as a plurality of individually controlled genes, as a polycistronic nucleic acid molecule, as a single nucleic acid molecule encoding a fusion protein, or any combination thereof.
  • endogenous or “native” refers to a polynucleotide, gene, protein, compound, molecule, or activity that is normally present in a host cell or a subject.
  • expression refers to the process by which a polypeptide is produced based on the encoding sequence of a nucleic acid molecule, such as a gene.
  • the process may include transcription, post-transcriptional control, post-transcriptional modification, translation, post-translational control, post- translational modification, or any combination thereof.
  • An expressed nucleic acid molecule is typically operably linked to an expression control sequence (e.g., a promoter).
  • operably linked refers to the association of two or more nucleic acid molecules on a single nucleic acid fragment so that the function of one is affected by the other.
  • a promoter is operably linked with a coding sequence when it is capable of affecting the expression of that coding sequence (i.e., the coding sequence is under the transcriptional control of the promoter).
  • Unlinked means that the associated genetic elements are not closely associated with one another and the function of one does not affect the other.
  • more than one heterologous nucleic acid molecule can be introduced into a host cell as separate nucleic acid molecules, as a plurality of individually controlled genes, as a polycistronic nucleic acid molecule, as a single nucleic acid molecule encoding a protein (e.g., a heavy chain of an antibody), or any combination thereof.
  • a protein e.g., a heavy chain of an antibody
  • two or more heterologous nucleic acid molecules are introduced into a host cell, it is understood that the two or more heterologous nucleic acid molecules can be introduced as a single nucleic acid molecule (e.g., on a single vector), on separate vectors, integrated into the host chromosome at a single site or multiple sites, or any combination thereof.
  • the number of referenced heterologous nucleic acid molecules or protein activities refers to the number of encoding nucleic acid molecules or the number of protein activities, not the number of separate nucleic acid molecules introduced into a host cell.
  • the term "construct” refers to any polynucleotide that contains a recombinant nucleic acid molecule (or, when the context clearly indicates, a fusion protein of the present disclosure).
  • a (polynucleotide) construct may be present in a vector (e.g., a bacterial vector, a viral vector) or may be integrated into a genome.
  • a "vector” is a nucleic acid molecule that is capable of transporting another nucleic acid molecule.
  • Vectors may be, for example, plasmids, cosmids, viruses, a RNA vector or a linear or circular DNA or RNA molecule that may include chromosomal, non-chromosomal, semi-synthetic or synthetic nucleic acid molecules.
  • Vectors of the present disclosure also include transposon systems (e.g., Sleeping Beauty, see, e.g., Geurts et al., Mol. Ther. 5:108, 2003: Mates et al., Nat. Genet. 41 :753, 2009).
  • transposon-based systems include, but are not limited to: piggybac (e.g., derived from lepidopteran cells and/or the Myotis lucifugus); mariner (e.g., derived from Drosophila); frog prince (e.g., derived from Rana pipiens); Tol2 (e.g., derived from medaka fish); and spinON.
  • Exemplary vectors are those capable of autonomous replication (episomal vector), capable of delivering a polynucleotide to a cell genome (e.g., viral vector), or capable of expressing nucleic acid molecules to which they are linked (expression vectors).
  • expression vector refers to a DNA construct containing a nucleic acid molecule that is operably linked to a suitable control sequence capable of effecting the expression of the nucleic acid molecule in a suitable host.
  • control sequences include a promoter to effect transcription, an optional operator sequence to control such transcription, a sequence encoding suitable mRNA ribosome binding sites, and sequences which control termination of transcription and translation.
  • the vector may be a plasmid, a phage particle, a virus, or simply a potential genomic insert.
  • the vector may replicate and function independently of the host genome, or may, in some instances, integrate into the genome itself or deliver the polynucleotide contained in the vector into the genome without the vector sequence.
  • plasmid "expression plasmid,” “virus,” and “vector” are often used interchangeably.
  • the term "introduced” in the context of inserting a nucleic acid molecule into a cell means “transfection", “transformation,” or “transduction” and includes reference to the incorporation of a nucleic acid molecule into a eukaryotic or prokaryotic cell wherein the nucleic acid molecule may be incorporated into the genome of a cell (e.g., chromosome, plasmid, plastid, or mitochondrial DNA), converted into an autonomous replicon, or transiently expressed (e.g., transfected mRNA).
  • a cell e.g., chromosome, plasmid, plastid, or mitochondrial DNA
  • transiently expressed e.g., transfected mRNA
  • polynucleotides of the present disclosure may be operatively linked to certain elements of a vector.
  • polynucleotide sequences that are needed to effect the expression and processing of coding sequences to which they are ligated may be operatively linked.
  • Expression control sequences may include appropriate transcription initiation, termination, promoter, and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (z.e., Kozak consensus sequences); sequences that enhance protein stability; and possibly sequences that enhance protein secretion.
  • Expression control sequences may be operatively linked if they are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
  • the vector comprises a plasmid vector or a viral vector (e.g., a lentiviral vector or a y-retroviral vector).
  • Viral vectors include retrovirus, adenovirus, parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as ortho-myxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses such as picornavirus and alphavirus, and double-stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus), and poxvirus (e.g., vaccinia, fowlpox, and canarypox).
  • herpesvirus e.
  • viruses include, for example, Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus.
  • retroviruses include avian leukosis-sarcoma, mammalian C-type, B-type viruses, D type viruses, HTLV-BLV group, lentivirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields et al., Eds., Lippincott-Raven Publishers, Philadelphia, 1996).
  • “Retroviruses” are viruses having an RNA genome, which is reverse-transcribed into DNA using a reverse transcriptase enzyme, the reverse-transcribed DNA is then incorporated into the host cell genome.
  • “Gammaretrovirus” refers to a genus of the retroviridae family. Examples of gammaretroviruses include mouse stem cell virus, murine leukemia virus, feline leukemia virus, feline sarcoma virus, and avian reticuloendotheliosis viruses.
  • Lentiviral vectors include HIV-based lentiviral vectors for gene delivery, which can be integrative or non-integrative, have relatively large packaging capacity, and can transduce a range of different cell types. Lentiviral vectors are usually generated following transient transfection of three (packaging, envelope, and transfer) or more plasmids into producer cells. Like HIV, lentiviral vectors enter the target cell through the interaction of viral surface glycoproteins with receptors on the cell surface. On entry, the viral RNA undergoes reverse transcription, which is mediated by the viral reverse transcriptase complex. The product of reverse transcription is a double-stranded linear viral DNA, which is the substrate for viral integration into the DNA of infected cells.
  • the viral vector can be a gammaretrovirus, e.g., Moloney murine leukemia virus (MLV)-derived vectors.
  • the viral vector can be a more complex retrovirus-derived vector, e.g., a lentivirus-derived vector. HIV-l-derived vectors belong to this category.
  • Other examples include lentivirus vectors derived from HIV-2, FIV, equine infectious anemia virus, SIV, and Maedi-Visna virus (ovine lentivirus).
  • Retroviral and lentiviral vector constructs and expression systems are also commercially available.
  • Other viral vectors also can be used for polynucleotide delivery including DNA viral vectors, including, for example adenovirus-based vectors and adeno-associated virus (AAV)-based vectors; vectors derived from herpes simplex viruses (HSVs), including amplicon vectors, replication-defective HSV and attenuated HSV (Krisky et al., Gene Ther. 5: 1517, 1998).
  • HSVs herpes simplex viruses
  • the viral vector may also comprise additional sequences between the two (or more) transcripts allowing for bicistronic or multi ci str onic expression.
  • additional sequences used in viral vectors include internal ribosome entry sites (IRES), furin cleavage sites, viral 2A peptide, or any combination thereof.
  • Plasmid vectors including DNA-based antibody or antigen-binding fragmentencoding plasmid vectors for direct administration to a subject, are described further herein.
  • the term "host” refers to a cell or microorganism targeted for genetic modification with a heterologous nucleic acid molecule to produce a polypeptide of interest (e.g., an antibody of the present disclosure).
  • a host cell may include any individual cell or cell culture which may receive a vector or the incorporation of nucleic acids or express proteins. The term also encompasses progeny of the host cell, whether genetically or phenotypically the same or different. Suitable host cells may depend on the vector and may include mammalian cells, animal cells, human cells, simian cells, insect cells, yeast cells, and bacterial cells. These cells may be induced to incorporate the vector or other material by use of a viral vector, transformation via calcium phosphate precipitation, DEAE-dextran, electroporation, microinjection, or other methods. See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual 2d ed. (Cold Spring Harbor Laboratory, 1989).
  • a "host” refers to a cell or a subject infected with a parvovirus.
  • a host subject is infected with parvovirus B 19 and/or has a chronic parvovirus infection.
  • Antigen refers to an immunogenic molecule that provokes an immune response. This immune response may involve antibody production, activation of specific immunologically-competent cells, activation of complement, antibody dependent cytotoxicicity, or any combination thereof.
  • An antigen immunogenic molecule
  • An antigen may be, for example, a peptide, glycopeptide, polypeptide, glycopolypeptide, polynucleotide, polysaccharide, lipid, or the like. It is readily apparent that an antigen can be synthesized, produced recombinantly, or derived from a biological sample. Exemplary biological samples that can contain one or more antigens include tissue samples, stool samples, cells, biological fluids, or combinations thereof.
  • Antigens can be produced by cells that have been modified or genetically engineered to express an antigen. Antigens can also be present in a parvovirus antigen, such as present in a virion, or expressed or presented on the surface of a cell infected by the parvovirus.
  • epitope includes any molecule, structure, amino acid sequence, or protein determinant that is recognized and specifically bound by a cognate binding molecule, such as an immunoglobulin, or other binding molecule, domain, or protein.
  • Epitopic determinants generally contain chemically active surface groupings of molecules, such as amino acids or sugar side chains, and can have specific three-dimensional structural characteristics, as well as specific charge characteristics.
  • the epitope can be comprised of consecutive amino acids (e.g., a linear epitope), or can be comprised of amino acids from different parts or regions of the protein that are brought into proximity by protein folding (e.g., a discontinuous or conformational epitope), or non-contiguous amino acids that are in close proximity irrespective of protein folding.
  • a linear epitope e.g., a linear epitope
  • protein folding e.g., a discontinuous or conformational epitope
  • non-contiguous amino acids that are in close proximity irrespective of protein folding.
  • the present disclosure provides an anti-parvovirus antibody, or an antigen-binding fragment thereof.
  • the parvovirus comprises a parvovirus Bl 9.
  • the parvovirus is a parvovirus Bl 9.
  • an antibody or antigen-binding fragment binds an epitope in B19 uVPl.
  • an antibody or antigen-binding fragment binds an epitope in B19 VP2.
  • the parvovirus human pathogen B 19 comprises an icosahedral nonenveloped capsid comprising VP1 (reported MW of 82 kDa) and VP2 (reported MW of 60 kDa) proteins.
  • VP1 comprises the entire sequence of VP2 coat protein, as well as a unique N-terminal region (uVPl or VPlu). See, e.g., Bilkova et al. Viruses 6(7):2899- 2937 (2014). Unless the context provides otherwise, a “VP 1 -specific” antibody or antigen-binding fragment binds to an epitope in uVPl.
  • an antibody or antigen-binding fragment of the present disclosure associates with or unites with a parvovirus (e.g., a B19, optionally binding to a uVPl or a VP2) while not significantly associating or uniting with any other molecules or components in a sample.
  • a parvovirus e.g., a B19, optionally binding to a uVPl or a VP2
  • an antibody or antigen-binding fragment of the present disclosure specifically binds to a uVPl or a VP2.
  • “specifically binds” refers to an association or union of an antibody or antigen-binding fragment to an antigen with an affinity or K a (i.e., an equilibrium association constant of a particular binding interaction with units of 1/M) equal to or greater than 10 5 M' 1 (which equals the ratio of the on-rate [K on ] to the off rate [K O ff] for this association reaction), while not significantly associating or uniting with any other molecules or components in a sample.
  • K a i.e., an equilibrium association constant of a particular binding interaction with units of 1/M
  • affinity may be defined as an equilibrium dissociation constant (Ka) of a particular binding interaction with units of M e.g., 10' 5 M to 10' 13 M).
  • Antibodies may be classified as “high-affinity” antibodies or as “low-affinity” antibodies.
  • “High-affinity” antibodies refer to those antibodies having a K a of at least 10 7 M -1 , at least 10 8 M' 1 , at least 10 9 M' 1 , at least IO 10 M' 1 , at least 10 11 M' 1 , at least 10 12 M' 1 , or at least 10 13 M' 1 .
  • “Low-affinity” antibodies refer to those antibodies having a K a of up to 10 7 M -1 , up to 10 6 M' 1 , up to 10 5 M' 1 .
  • affinity may be defined as an equilibrium dissociation constant (Kd) of a particular binding interaction with units of M (e.g., 10' 5 M to 10' 13 M).
  • assays for identifying antibodies of the present disclosure that bind a particular target, as well as determining binding domain or binding protein affinities, such as Western blot, ELISA (e.g., direct, indirect, or sandwich), analytical ultracentrifugation, spectroscopy, biolayer interferometry, and surface plasmon resonance (Biacore®) analysis (see, e.g., Scatchard et al., Ann. N.Y. Acad. Sci. 57:660, 1949; Wilson, Science 295:2103, 2002; Wolff et al., Cancer Res. 53:2560, 1993; and U.S. Patent Nos. 5,283,173, 5,468,614, or the equivalent). Assays for assessing affinity or apparent affinity or relative affinity are also known.
  • binding can be determined by recombinantly expressing a uVPl or a VP2 in a host cell (e.g., by transfection) and immunostaining the (e.g, fixed, or fixed and permeabilized) host cell with antibody and analyzing binding by flow cytometery (e.g., using a ZE5 Cell Analyzer (BioRad®) and FlowJo software (TreeStar).
  • positive binding can be defined by differential staining by antibody of uVPl - or VP2-expressing cells versus control (e.g., mock) cells.
  • an antibody or antigen-binding fragment of the present disclosure binds to a uVPl or a VP2, as measured using biolayer interferometry, or by surface plasmon resonance, or by ELISA.
  • the IC50 is the concentration of a composition (e.g., antibody) that results in half-maximal inhibition of the indicated biological or biochemical function, activity, or response.
  • the EC50 is the concentration of a composition that provides the half-maximal response in the assay.
  • IC50 and EC50 are used interchangeably.
  • an antibody of the present disclosure is capable of neutralizing infection by a parvovirus.
  • a “neutralizing antibody” is one that can neutralize, i.e., prevent, inhibit, reduce, impede, or interfere with, the ability of a pathogen to initiate and/or perpetuate an infection in a host.
  • the terms "neutralizing antibody” and “an antibody that neutralizes” or “antibodies that neutralize” are used interchangeably herein.
  • the antibody or antigen-binding fragment can be capable of preventing and/or neutralizing a parvovirus infection in an in vitro model of infection and/or in an in vivo animal model of infection and/or in a human.
  • the antibody, or antigen-binding fragment thereof is human, humanized, or chimeric.
  • antibody refers to an intact antibody comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, as well as any antigen-binding portion or fragment of an intact antibody that has or retains the ability to bind to the antigen target molecule recognized by the intact antibody, such as an scFv, Fab, or Fab'2 fragment.
  • antibody herein is used in the broadest sense and includes polyclonal and monoclonal antibodies, including intact antibodies and functional (antigen-binding) antibody fragments thereof, including fragment antigen binding (Fab) fragments, F(ab')2 fragments, Fab' fragments, Fv fragments, recombinant IgG (rlgG) fragments, single chain antibody fragments, including single chain variable fragments (scFv), and single domain antibodies (e.g., sdAb, sdFv, nanobody) fragments.
  • Fab fragment antigen binding
  • rlgG recombinant IgG
  • scFv single chain variable fragments
  • single domain antibodies e.g., sdAb, sdFv, nanobody
  • the term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intrabodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies, and heteroconjugate antibodies, multi specific, e.g., bi specific antibodies, diabodies, triabodies, tetrabodies, tandem di-scFv, and tandem tri-scFv.
  • antibody should be understood to encompass functional antibody fragments thereof.
  • the term also encompasses intact or full-length antibodies, including antibodies of any class or sub-class, including IgG and sub-classes thereof (IgGl, IgG2, IgG3, IgG4), IgM, IgE, IgA, and IgD.
  • VL or “VL” and “ VH” or “VH” refer to the variable binding region from an antibody light chain and an antibody heavy chain, respectively.
  • a VL is a kappa (K) class (also “VK” herein).
  • a VL is a lambda (X) class.
  • the variable binding regions comprise discrete, well-defined sub-regions known as “complementarity determining regions” (CDRs) and “framework regions” (FRs).
  • CDR complementarity determining region
  • HVR hypervariable region
  • an antibody VH comprises four FRs and three CDRs as follows: FR1-HCDR1-FR2-HCDR2-FR3-HCDR3-FR4; and an antibody VL comprises four FRs and three CDRs as follows: FR1-LCDR1-FR2- LCDR2-FR3-LCDR3-FR4.
  • the VH and the VL together form the antigenbinding site through their respective CDRs.
  • one or more CDRs do not contact antigen and/or do not contribute energetically to antigen binding.
  • a "variant" of a CDR refers to a functional variant of a CDR sequence having up to 1-3 amino acid substitutions (e.g., conservative or nonconservative substitutions), deletions, or combinations thereof.
  • a functional variant of a CDR substantially retains the conformation and optionally the binding function of a native or parent CDR.
  • Numbering of CDR and framework regions may be according to any known method or scheme, such as the Kabat, Chothia, EU, IMGT, Contact, North, Martin, AbM, and AHo numbering schemes see, e.g., Kabat et al., "Sequences of Proteins of Immunological Interest, US Dept. Health and Human Services, Public Health Service National Institutes of Health, 1991, 5 th ed.; Chothia and Lesk, J. Mol. Biol. 796:901-917 (1987)); Lefranc et al., Dev. Comp. Immunol. 27:55, 2003; Honegger and Pliickthun, J. Mol. Bio. 309:65'1-6'10 (2001); North et al. J Mol Biol. (2011) 406:228-56; doi: 10.1016/j.jmb.2010.10.030; Abhinandan and Martin, Mol
  • an antibody or antigen-binding fragment comprises one or more CDRs of a VH sequence according to any one of SEQ ID NOs.:7, 17, and 27, and/or of a VL sequence according to any one of SEQ ID NOs.: 12, 22, and 32, in accordance with any known CDR numbering method, including the Kabat, Chothia, North, EU, IMGT, Martin (Enhanced Chothia), Contact, AbM, and AHo numbering methods, or in accordance with a combination of two or more of these methods (e.g., including those residues falling within a CDR as defined by either or all of the numbering methods).
  • CDR numbering method including the Kabat, Chothia, North, EU, IMGT, Martin (Enhanced Chothia), Contact, AbM, and AHo numbering methods, or in accordance with a combination of two or more of these methods (e.g., including those residues falling within a CDR as defined by either or all of the numbering methods).
  • CDRs are according to the IMGT numbering method (optionally using junction definitions for CDR3 amino acid sequences).
  • CDRs are according to the antibody numbering method developed by the Chemical Computing Group (CCG); e.g., using Molecular Operating Environment (MOE) software (chemcomp.com).
  • CDRs are in accordance with the IMGT numbering method.
  • an antibody or antigen-binding fragment comprises a CDRH1, a CDRH2, a CDRH3, a CDRL1, a CDRL2, and/or a CDRL3 of exemplary antibody PAB18, PAB5, or PAA19, as set forth in Table A.
  • an antibody or antigen-binding fragment comprises a VH and/or a VL of exemplary antibody PAB18, PAB5, or PAA19, as set forth in Table A.
  • Variable domain amino acid sequences are also shown for antibodies PAB18, PAB5, and PAA19 in Figures 8A-8C, respectively.
  • Table B shows certain variable domain germline alleles corresponding to PAB18, PAB5, and PAA19, and percent identity of the antibody VH domain to the VH germline sequence.
  • an antibody or antigen-binding fragment comprises a VH comprising an amino acid sequence having at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% to the amino acid sequence encoded by VH1-69.
  • the antibody or antigen-binding fragment comprises a VL of kappa class.
  • an antibody or antigen-binding fragment comprises a VH comprising an amino acid sequence having at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% to the amino acid sequence encoded by VH3-21.
  • the antibody or antigen-binding fragment comprises a VL of kappa class.
  • an antibody or antigen-binding fragment comprises a VH comprising an amino acid sequence having at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% to the amino acid sequence encoded by VH4-39.
  • the antibody or antigen-binding fragment comprises a VL of kappa class.
  • an antibody or antigen-binding fragment comprises the CDRH3 amino acid sequence of the VH amino acid sequence set forth in SEQ ID NO.:7 and the CDRL3 amino acid sequence of the VL amino acid sequence set forth in SEQ ID NO. : 12, wherein the CDRH3 and the CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an antibody or antigen-binding fragment comprises the CDRH3 amino acid sequence of the VH amino acid sequence set forth in SEQ ID NO. : 17 and the CDRL3 amino acid sequence of the VL amino acid sequence set forth in SEQ ID NO.: 22, wherein the CDRH3 and CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an antibody or antigen-binding fragment comprises the CDRH3 amino acid sequence of the VH amino acid sequence set forth in SEQ ID NO.:27 and the CDRL3 amino acid sequence of the VL amino acid sequence set forth in SEQ ID NO.: 32, wherein the CDRH3 and CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an antibody or antigen-binding fragment comprises the CDRH1, CDRH2, and CDRH3 amino acid sequences of the VH amino acid sequence set forth in SEQ ID NO.:7, and the CDRL1, CDRL2, and CDRL3 amino acid sequences of the VL amino acid sequences set forth in SEQ ID NO.: 12, wherein the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an antibody or antigen-binding fragment comprises the CDRH1, CDRH2, and CDRH3 amino acid sequences of the VH amino acid sequence set forth in SEQ ID NO.: 17, and the CDRL1, CDRL2, and CDRL3 amino acid sequences of the VL amino acid sequences set forth in SEQ ID NO.:22, wherein the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an antibody or antigen-binding fragment comprises the CDRH1, CDRH2, and CDRH3 amino acid sequences of the VH amino acid sequence set forth in SEQ ID NO.:27, and the CDRL1, CDRL2, and CDRL3 amino acid sequences of the VL amino acid sequences set forth in SEQ ID NO.:32, wherein the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 are according to IMGT, or are according to Kabat, or are according to Chothia, or are according to Martin, or are according to AHo, or are according to North, or are according to AbM, or are according to CCG, or are according to Contact, or are according to EU, or are according to a combination of two or more of the foregoing.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)Hl, the CDRH2, and the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.: 17; and (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:22, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment of the present disclosure binds to an epitope in B19 uVPl. In certain embodiments, an anti-parvovirus antibody or antigen-binding fragment binds to a peptide having the amino acid sequence TGTDLE (SEQ ID NO.:4).
  • an antiparvovirus antibody or antigen-binding fragment binds to a peptide having the amino acid sequence X1X2TDX3EX4 (SEQ ID NO.: 5), wherein: Xi is present or absent, and, if present, is any amino acid; X2 is G or F, and is preferably G; X3 is L or K, and is preferably K; and X4 is present or absent, and, if present, is any amino acid except P.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, and/or the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.: 17; and/or (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:22, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)Hl, the CDRH2, and the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.: 17; and (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:22, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H3 amino acid sequence set forth in SEQ ID NO.:20, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; and (ii) in a/the light chain variable domain (VL), the complementarity determining region (CDR)L3 amino acid sequence set forth in SEQ ID NO.:25, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H1 amino acid sequence set forth in SEQ ID NO. : 18, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the complementarity determining region (CDR)H2 amino acid sequence set forth in SEQ ID NO.: 19, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; and (ii) in a/the light chain variable domain (VL), the CDRL1 amino acid sequence set forth in SEQ ID NO.:23, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the CDRL2 amino acid sequence set forth in SEQ ID NO.:24, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • VH heavy chain variable domain
  • CDR complementarity determining
  • an anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.: 18-20 and 23-25, respectively.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.: 17; and/or (ii) a light chain variable domain (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.:22.
  • amino acids amino acids having at least
  • Framework regions can be identified according to a numbering scheme (e.g., IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo, or a combination of two or more of these).
  • the CDRs may be identified within a variable domain or within a heavy or light chain according to a numbering scheme or a combination of numbering schemes, and, preferably, the FRs may be identified using the same numbering scheme or combination of numbering schemes.
  • an anti-parvovirus antibody or antigen-binding fragment comprises a VH comprising a FR1, a FR2, a FR3, and/or a FR4 (or a variant of the FR1, FR2, FR3, and/or FR4 comprising one, two, three, four, or five amino acid substitutions, insertions, and/or deletions) of the VH amino acid sequence set forth in SEQ ID NO.
  • VL comprising a a FR1, a FR2, a FR3, and/or a FR4 (or a variant of the FR1, FR2, FR3, and/or FR4 comprising one, two, three, four, or five amino acid substitutions, insertions, and/or deletions) of the VL amino acid sequence set forth in SEQ ID NO.:22.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO.: 17; and (ii) a light chain variable domain (VL) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO.:22.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising the amino acid sequence set forth in SEQ ID NO.: 17; and (ii) a light chain variable domain (VL) comprising the amino acid sequence set forth in SEQ ID NO.:22.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) consisting essentially of the amino acid sequence set forth in SEQ ID NO.: 17; and (ii) a light chain variable domain (VL) consisting essentially of the amino acid sequence set forth in SEQ ID NO.:22.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) consisting of the amino acid sequence set forth in SEQ ID NO.: 17; and (ii) a light chain variable domain (VL) consisting of the amino acid sequence set forth in SEQ ID NO.:22.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment binds to an epitope in B19 VP2.
  • an antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, and/or the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:7; and/or (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.: 12, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an antiparvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region CDR)H1, the CDRH2, the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:7; and in a light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.: 12, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, and/or the CDRH3 of the VH amino acid sequence set forth in SEQ ID N0.:7; and/or (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.: 12, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region CDR)H1, the CDRH2, the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:7; and in a light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H3 amino acid sequence set forth in SEQ ID NO.: 10, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; and (ii) in a/the light chain variable domain (VL), the complementarity determining region (CDR)L3 amino acid sequence set forth in SEQ ID NO.: 15, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H1 amino acid sequence set forth in SEQ ID NO.:8, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the complementarity determining region (CDR)H2 amino acid sequence set forth in SEQ ID NO.:9, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; and (ii) in a/the light chain variable domain (VL), the CDRL1 amino acid sequence set forth in SEQ ID NO.: 13, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the CDRL2 amino acid sequence set forth in SEQ ID NO. : 14, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • an anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.:8-10 and 13-15, respectively.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.:7; and/or (ii) a light chain variable domain (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.: 12.
  • VH heavy chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises a VH comprising a FR1, a FR2, a FR3, and/or a FR4 (or a variant of the FR1, FR2, FR3, and/or FR4 comprising one, two, three, four, or five amino acid substitutions, insertions, and/or deletions) of the VH amino acid sequence set forth in SEQ ID NO.
  • VL comprising a a FR1, a FR2, a FR3, and/or a FR4 (or a variant of the FR1, FR2, FR3, and/or FR4 comprising one, two, three, four, or five amino acid substitutions, insertions, and/or deletions) of the VL amino acid sequence set forth in SEQ ID NO. : 12.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO.:7; and (ii) a light chain variable domain (VL) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO. : 12.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising the amino acid sequence set forth in SEQ ID NO.:7; and (ii) a light chain variable domain (VL) comprising the amino acid sequence set forth in SEQ ID NO. : 12.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) consisting essentially of the amino acid sequence set forth in SEQ ID NO.:7; and (ii) a light chain variable domain (VL) consisting essentially of the amino acid sequence set forth in SEQ ID NO.: 12.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, and/or the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:27; and/or (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment of comprises: (i) in a heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, and/or the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:27; and/or (ii) in a light chain variable domain (VL), the CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:27; and (ii) in a/the light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H1, the CDRH2, the CDRH3 of the VH amino acid sequence set forth in SEQ ID NO.:27; and (ii) in a/the light chain variable domain (VL), the CDRL1, the CDRL2, and the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H3 amino acid sequence set forth in SEQ ID NO.:30, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; (ii) in a/the light chain variable domain (VL), the complementarity determining region (CDR)L3 amino acid sequence set forth in SEQ ID NO.:35, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • an anti-parvovirus antibody or antigenbinding fragment comprises: (i) in a/the heavy chain variable domain (VH), the complementarity determining region (CDR)H1 amino acid sequence set forth in SEQ ID NO.:28, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the complementarity determining region (CDR)H2 amino acid sequence set forth in SEQ ID NO.:29, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions; and (ii) in a/the light chain variable domain (VL), the CDRL1 amino acid sequence set forth in SEQ ID NO.:33, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions, and the CDRL2 amino acid sequence set forth in SEQ ID NO.:34, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • VH heavy chain variable domain
  • CDR complementarity determining
  • an anti-parvovirus antibody or antigen-binding fragment thereof comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.:28-30 and 33-35, respectively.
  • an anti-parvovirus antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable domain (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.:27; and/or (ii) a light chain variable domain (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.:32.
  • VH heavy chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises a VH comprising a FR1, a FR2, a FR3, and/or a FR4 (or a variant of the FR1, FR2, FR3, and/or FR4 comprising one, two, three, four, or five amino acid substitutions, insertions, and/or deletions) of the VH amino acid sequence set forth in SEQ ID NO.
  • the FRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO.:27; and (ii) a light chain variable domain (VL) comprising, consisting essentially of, or consisting of the amino acid sequence set forth in SEQ ID NO.:32.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) comprising the amino acid sequence set forth in SEQ ID NO.:27; and (ii) a light chain variable domain (VL) comprising the amino acid sequence set forth in SEQ ID NO.:32.
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) consisting essentially of the amino acid sequence set forth in SEQ ID NO.:27; and (ii) a light chain variable domain (VL) consisting essentially of the amino acid sequence set forth in SEQ ID NO.:32.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • an anti-parvovirus antibody or antigen-binding fragment comprises: (i) a heavy chain variable domain (VH) consisting of the amino acid sequence set forth in SEQ ID NO.:27; and (ii) a light chain variable domain (VL) consisting of the amino acid sequence set forth in SEQ ID NO.:32.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • CL refers to an "immunoglobulin light chain constant region” or a "light chain constant region,” z.e., a constant region from an antibody light chain.
  • CH refers to an "immunoglobulin heavy chain constant region” or a "heavy chain constant region,” which is further divisible, depending on the antibody isotype, into CHI, CH2, and CH3 (IgA, IgD, IgG), or CHI, CH2, CH3, and CH4 domains (IgE, IgM).
  • CHI unimmunoglobulin heavy chain constant region
  • an antibody or antigen-binding fragment of the present disclosure comprises any one or more of CL, a CHI, a CH2, and a CH3.
  • an antibody or antigen-binding fragment of the present disclosure may comprise any one or more of CL, a CHI, a CH2, and a CH3.
  • a CL comprises an amino acid sequence having at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the amino acid sequence of SEQ ID NO.:36.
  • a CL comprises an amino acid sequence having at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to a human lambda light chain constant domain.
  • a CH1-CH3 comprises an amino acid sequence having at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the amino acid sequence of any one of SEQ ID NOs.:38-42.
  • a Fc polypeptide comprises an amino acid sequence having at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the amino acid sequence of SEQ ID NO.:37.
  • an antibody or antigen-binding fragment of the present disclosure can comprise a heavy chain, a CH1-CH3, a CH3, or an Fc polypeptide wherein a C-terminal lysine residue is present or is absent; in other words, encompassed are embodiments where the C-terminal residue of a heavy chain, a CH1- CH3, or an Fc polypeptide is not a lysine, and embodiments where a lysine is the C- terminal residue.
  • a composition comprises a plurality of an antibody and/or an antigen-binding fragment of the present disclosure, wherein one or more antibody or antigen-binding fragment does not comprise a lysine residue at the C- terminal end of the heavy chain, CH1-CH3, or Fc polypeptide, and wherein one or more antibody or antigen-binding fragment comprises a lysine residue at the C-terminal end of the heavy chain, CH1-CH3, or Fc polypeptide.
  • AFc, Fc fragment or portion, or antibody may be of any allotype or combination of allotypes.
  • Allotype refers to the allelic variation found among the IgG subclasses.
  • an allotype may comprise Glml (or Glm(a)), Glm2 (or Glm(x)), Glm3 (or Glm(f)), Glml7 (or Gm(z))m), Glm27, and/or Glm28 (Glm27 and Glm28 have been described as “alloallotypes”).
  • the Glm3 and Glml7 allotypes are located at the same position in the CHI domain (position 214 according to EU numbering).
  • Glm3 comprises R214 (EU)
  • Glml7 comprises K214 (EU).
  • the Glml allotype is located in the CH3 domain (at positions 356 and 358 (EU)) and refers to the replacements E356D and M358L.
  • the Glm2 allotype refers to a replacement of the alanine in position 431 (EU) by a glycine.
  • Glm allotypes, alloallotypes, and features thereof are known in the art and described at, for example, www.imgt.org/IMGTrepertoire/Proteins/allotypes/human/IGH/IGHC/Glm_allotypes.ht ml and Lefranc, M.-P. and Lefranc, G. Human Gm, Km and Am allotypes and their molecular characterization: a remarkable demonstration of polymorphism In: B. Tait, E Christiansen (Eds.), Immunogenetics, chap. 34, Humana Press, Springer, New York, USA. Methods Mol. Biol. 2012; 882, 635-680. PMID: 22665258, LIGM: 406, the contents and allotypes and allotype information of which are incorporated herein by reference.
  • the Glml allotype may be combined, for example, with the Glm3, Glm 17, Glm27, Glm2, and/or Glm28 allotype.
  • an allotype is Glm3 with no Glml (Glm3,-1).
  • an allotype is Glml7,l allotype.
  • an allotype is Glm3,l.
  • an allotype is Glml7 with no Glml (Glml7,-1).
  • these allotypes may be combined (or not combined) with the Glm2, Glm27 or Glm28 allotype.
  • an allotype may be Glml7,l,2.
  • a polypeptide, CH2, Fc, Fc fragment or portion, or antibody of the present disclosure comprises a Glm3 allotype or a Glm3,l allotype.
  • a polypeptide, CH2, Fc, Fc fragment or portion, or antibody of the present disclosure comprises a Glm3 allotype and comprises M428L and N434S or M428L and N434A mutations or any other mutation(s) that enhance binding to a human FcRn, such as those described herein.
  • a polypeptide, CH2, Fc, Fc fragment or portion, or antibody of the present disclosure comprises a Glm3,l allotype and comprises M428L and N434S or M428L and N434A mutations or any other mutation(s) that enhance binding to a human FcRn, such as those described herein.
  • a polypeptide, CH2, Fc, Fc fragment or portion, or antibody of the present disclosure comprises a Glml7, 1 allotype.
  • a polypeptide, CH2, Fc, Fc fragment or portion, or antibody of the present disclosure comprises a Glml7, 1 allotype and comprises M428L and N434S or M428L and N434A mutations or any other mutation(s) that enhance binding to a human FcRn, as described further herein.
  • a “Fab” fragment antigen binding is the part of an antibody that binds to antigens and includes the variable region and CHI of the heavy chain linked to the light chain via an inter-chain disulfide bond. Each Fab fragment is monovalent with respect to antigen binding, z.e., it has a single antigen-binding site. Pepsin treatment of an antibody yields a single large F(ab')2 fragment that roughly corresponds to two disulfide linked Fab fragments having divalent antigen-binding activity and is still capable of cross-linking antigen.
  • Both the Fab and F(ab’)2 are examples of "antigenbinding fragments.”
  • Fab' fragments differ from Fab fragments by having additional few residues at the carboxy terminus of the CHI domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • Fab fragments may be joined, e.g., by a peptide linker, to form a single chain Fab, also referred to herein as "scFab.”
  • a single chain Fab also referred to herein as "scFab.”
  • an inter-chain disulfide bond that is present in a native Fab may not be present, and the linker serves in full or in part to link or connect the Fab fragments in a single polypeptide chain.
  • a heavy chain- derived Fab fragment e.g., comprising, consisting of, or consisting essentially of VH + CHI, or "Fd
  • a light chain-derived Fab fragment e.g., comprising, consisting of, or consisting essentially of VL + CL
  • a scFab may be arranged, in N-terminal to C-terminal direction, according to (heavy chain Fab fragment - linker - light chain Fab fragment) or (light chain Fab fragment - linker - heavy chain Fab fragment).
  • Peptide linkers and exemplary linker sequences for use in scFabs are discussed in further detail herein.
  • Fv is a small antibody fragment that contains a complete antigen-recognition and antigen-binding site. This fragment generally consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although typically at a lower affinity than the entire binding site.
  • Single-chain Fv also abbreviated as “sFv” or “scFv”
  • sFv single-chain Fv
  • the scFv polypeptide comprises a polypeptide linker disposed between and linking the VH and VL domains that enables the scFv to retain or form the desired structure for antigen binding.
  • a peptide linker can be incorporated into a fusion polypeptide using standard techniques well known in the art.
  • the antibody or antigen-binding fragment comprises a scFv comprising a VH domain, a VL domain, and a peptide linker linking the VH domain to the VL domain.
  • a scFv comprises a VH domain linked to a VL domain by a peptide linker, which can be in a VH-linker- VL orientation or in a VL-linker-VH orientation.
  • Any scFv of the present disclosure may be engineered so that the C-terminal end of the VL domain is linked by a short peptide sequence to the N-terminal end of the VH domain, or vice versa (i.e., (N)VL(C)-linker-(N)VH(C) or (N)VH(C)-linker-(N)VL(C).
  • a linker may be linked to an N-terminal portion or end of the VH domain, the VL domain, or both.
  • Peptide linker sequences may be chosen, for example, based on: (1) their ability to adopt a flexible extended conformation; (2) their inability or lack of ability to adopt a secondary structure that could interact with functional epitopes on the first and second polypeptides and/or on a target molecule; and/or (3) the lack or relative lack of hydrophobic or charged residues that might react with the polypeptides and/or target molecule.
  • linker design e.g., length
  • linker design can include the conformation or range of conformations in which the VH and VL can form a functional antigen-binding site.
  • peptide linker sequences contain, for example, Gly, Asn and Ser residues.
  • linker sequence may also be included in a linker sequence.
  • Other amino acid sequences which may be usefully employed as linker include those disclosed in Maratea et al., Gene 40:39 46 (1985); Murphy et al., Proc. Natl. Acad. Sci. USA 83:8258 8262 (1986); U.S. Pat. No. 4,935,233, and U.S. Pat. No. 4,751,180.
  • linkers may include, for example, Glu-Gly-Lys-Ser-Ser-Gly-Ser-Gly-Ser-Glu-Ser-Lys- Val-Asp (Chaudhary et al., Proc. Natl. Acad. Sci.
  • Any suitable linker may be used, and in general can be about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 15 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100 amino acids in length, or less than about 200 amino acids in length, and will preferably comprise a flexible structure (can provide flexibility and room for conformational movement between two regions, domains, motifs, fragments, or modules connected by the linker), and will preferably be biologically inert and/or have a low risk of immunogenicity in a human.
  • ScFvs can be constructed using any combination of the VH and VL sequences or any combination of the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 sequences disclosed herein.
  • linker sequences are not required; for example, when the first and second polypeptides have non-essential N-terminal amino acid regions that can be used to separate the functional domains and prevent steric interference.
  • DNA in the germline variable (V), joining (J), and diversity (D) gene loci may be rearranged and insertions and/or deletions of nucleotides in the coding sequence may occur. Somatic mutations may be encoded by the resultant sequence, and can be identified by reference to a corresponding known germline sequence.
  • somatic mutations that are not critical to a desired property of the antibody e.g., binding to a uVPl or VP2 polypeptide
  • that confer an undesirable property upon the antibody e.g., an increased risk of immunogenicity in a subject administered the antibody
  • the antibody or antigen-binding fragment of the present disclosure comprises at least one more germline-encoded amino acid in a variable region as compared to a parent antibody or antigen-binding fragment, provided that the parent antibody or antigen binding fragment comprises one or more somatic mutations.
  • Variable region and CDR amino acid sequences of exemplary anti-parvovirus antibodies of the present disclosure are provided in Table 1 herein.
  • IMGT.org see e.g. www. imgt. org/IMGT_vquest/ analy si s).
  • an antibody or antigen-binding fragment comprises an amino acid modification (e.g., a substitution mutation) to remove an undesired risk of oxidation, deamidation, and/or isomerization.
  • an amino acid modification e.g., a substitution mutation
  • variant antibodies that comprise one or more amino acid alterations in a variable region (e.g., VH, VL, framework or CDR) as compared to a presently disclosed (“parent”) antibody, wherein the variant antibody is capable of binding to a parvovirus antigen.
  • a variable region e.g., VH, VL, framework or CDR
  • composition or combination that comprises a first antibody or antigen-binding fragment, and a different, second antibody or antigen-binding fragment, wherein the first antibody or antigen-binding fragment binds to an epitope in uVPl and the second antibody or antigen-binding fragment binds to an epitope in VP2.
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigenbinding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A see also Figure 8A).
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A see also Figure 8B), and the second antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18, as shown in Table A (see also Figure 8 A).
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • an antibody or antigen-binding fragment of the present disclosure is monospecific (e.g., binds to a single epitope) or is multispecific (e.g., binds to multiple epitopes and/or target molecules).
  • Antibodies and antigen binding fragments may be constructed in various formats. Exemplary antibody formats disclosed in Spiess et al., Mol. Immunol.
  • FIT-Ig e.g., PCT Publication No.
  • WuxiBody formats e.g., PCT Publication No. WO 2019/057122, which formats are incorporated herein by reference in their entirety
  • In-Elbow-Insert Ig formats lELIg; e.g., PCT Publication Nos. WO 2019/024979 and WO 2019/025391, which formats are incorporated herein by reference in their entirety.
  • the antibody or antigen-binding fragment comprises two or more of VH domains, two or more VL domains, or both (z.e., two or more VH domains and two or more VL domains).
  • an antigen-binding fragment comprises the format (N-terminal to C-terminal direction) VH-linker- VL- linker-VH-linker-VL, wherein the two VH sequences can be the same or different and the two VL sequences can be the same or different.
  • Such linked scFvs can include any combination of VH and VL domains arranged to bind to a given target, and in formats comprising two or more VH and/or two or more VL, one, two, or more different eptiopes or antigens may be bound. It will be appreciated that formats incorporating multiple antigen-binding domains may include VH and/or VL sequences in any combination or orientation.
  • the antigen-binding fragment can comprise the format VL-linker-VH-linker-VL-linker-VH, VH-linker-VL-linker-VL-linker-VH, or VL-linker- VH-linker- VH-linker- VL .
  • Monospecific or multispecific antibodies or antigen-binding fragments of the present disclosure constructed comprise any combination of the VH and VL sequences and/or any combination of the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 sequences disclosed herein.
  • a bispecific or multispecific antibody or antigenbinding fragment may, in some embodiments, comprise one, two, or more antigenbinding domains (e.g., a VH and a VL) of the instant disclosure.
  • Two or more binding domains may be present that bind to the same or a different epitope, and a bispecific or multispecific antibody or antigen-binding fragment as provided herein can, in some embodiments, comprise a further parvovirus-specific binding domain, and/or can comprise a binding domain that binds to a different antigen or pathogen altogether.
  • the antibody or antigen-binding fragment can be multispecific; e.g., bispecific, trispecific, or the like.
  • a multispecific antibody or antigen-binding fragment is provided that comprises the heavy and light chain CDRs of PAB5 and PAB18; or of PAB5 and PAA19.
  • multispecific antibodies or antigen-binding fragments comprising the VH and the VL of PAB5 and PAB18; or of P AB 5 and PAA19; or of PAB18 and PAA19.
  • the multispecific antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • the antibody or antigen-binding fragment comprises a Fc polypeptide, or a fragment thereof.
  • the "Fc" fragment or Fc polypeptide comprises the carboxy -terminal portions (ie., the CH2 and CH3 domains of IgG) of both antibody H chains held together by disulfides.
  • An Fc may comprise a dimer comprised of two Fc polypeptides (z.e., two CH2-CH3 polypeptides).
  • Antibody "effector functions" refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype.
  • antibody effector functions include: Clq binding and complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor); and B cell activation.
  • modifications e.g., amino acid substitutions
  • Fc domain in order to modify (e.g., improve, reduce, or ablate) one or more functionality of an Fc-containing polypeptide (e.g., an antibody of the present disclosure).
  • Such functions include, for example, Fc receptor (FcR) binding, antibody half-life modulation (e.g., by binding to FcRn), ADCC function, protein A binding, protein G binding, and complement binding.
  • Amino acid modifications that modify (e.g., improve, reduce, or ablate) Fc functionalities include, for example, the T250Q/M428L, M252Y/S254T/T256E, H433K/N434F, M428L/N434S, M428L/N434A, E233P/L234V/L235A/G236 + A327G/A330S/P331S, E333A, S239D/A330L/I332E, P257I/Q311, K326W/E333S, S239D/I332E/G236A, N297Q, K322A, S228P, L235E + E318A/K320A/K322A, L234A/L235A (also referred to herein as "LALA"), and L234A/L235A/P329G mutations, which mutations are summarized and annotated in "Engineered Fc Regions
  • the Clq protein complex can bind to at least two molecules of IgGl or one molecule of IgM when the immunoglobulin molecule(s) is attached to the antigenic target (Ward, E. S., and Ghetie, V., Then Immunol. 2 (1995) 77-94).
  • Burton, D. R. described (Mol. Immunol. 22 (1985) 161-206) that the heavy chain region comprising amino acid residues 318 to 337 is involved in complement fixation.
  • Duncan, A. R., and Winter, G. (Nature 332 (1988) 738-740), using site directed mutagenesis, reported that Glu318, Lys320 and Lys322 form the binding site to Clq.
  • the role of Glu318, Lys320 and Lys 322 residues in the binding of Clq was confirmed by the ability of a short synthetic peptide containing these residues to inhibit complement mediated lysis.
  • FcR binding can be mediated by the interaction of the Fc moiety (of an antibody) with Fc receptors (FcRs), which are specialized cell surface receptors on cells including hematopoietic cells.
  • Fc receptors belong to the immunoglobulin superfamily, and shown to mediate both the removal of antibody-coated pathogens by phagocytosis of immune complexes, and the lysis of erythrocytes and various other cellular targets (e.g. tumor cells) coated with the corresponding antibody, via antibody dependent cell mediated cytotoxicity (ADCC; Van de Winkel, J. G., and Anderson, C. L., J. Leukoc. Biol. 49 (1991) 511-524).
  • ADCC antibody dependent cell mediated cytotoxicity
  • FcRs are defined by their specificity for immunoglobulin classes; Fc receptors for IgG antibodies are referred to as FcyR, for IgE as FcsR, for IgA as FcaR and so on and neonatal Fc receptors are referred to as FcRn.
  • Fc receptor binding is described for example in Ravetch, J. V, and Kinet, J. P, Annu. Rev. Immunol. 9 (1991) 457-492; Capel, P. J., et al., Immunomethods 4 (1994) 25-34; de Haas, M., et al., J Lab. Clin. Med. 126 (1995) 330-341; and Gessner, J. E., et al., Ann. Hematol. 76 (1998) 231-248.
  • FcyR Fc domain of native IgG antibodies
  • FcyR In humans, three classes of FcyR have been characterized to-date, which are: (i) FcyRI (CD64), which binds monomeric IgG with high affinity and is expressed on macrophages, monocytes, neutrophils and eosinophils; (ii) FcyRII (CD32), which binds complexed IgG with medium to low affinity, is widely expressed, in particular on leukocytes, is believed to be a central player in antibody-mediated immunity, and which can be divided into FcyRIIA, FcyRIIB and FcyRIIC, which perform different functions in the immune system, but bind with similar low affinity to the IgG-Fc, and the ectodomains of these receptors are highly homologuous; and (iii) FcyRIII (CD 16), which binds IgG with medium to low affinity and has been found in two forms: FcyRIIIA, which has been found on NK cells, macrophages,
  • FcyRIIA is found on many cells involved in killing (e.g. macrophages, monocytes, neutrophils) and seems able to activate the killing process.
  • FcyRIIB seems to play a role in inhibitory processes and is found on B-cells, macrophages and on mast cells and eosinophils. Importantly, it has been shown that 75% of all FcyRIIB is found in the liver (Ganesan, L. P. et al., 2012: “FcyRIIb on liver sinusoidal endothelium clears small immune complexes," Journal of Immunology 189: 4981-4988).
  • FcyRIIB is abundantly expressed on Liver Sinusoidal Endothelium, called LSEC, and in Kupffer cells in the liver and LSEC are the major site of small immune complexes clearance (Ganesan, L. P. et al., 2012: FcyRIIb on liver sinusoidal endothelium clears small immune complexes. Journal of Immunology 189: 4981-4988).
  • the antibodies disclosed herein and the antigen-binding fragments thereof comprise an Fc polypeptide or fragment thereof for binding to FcyRIIb, in particular an Fc region, such as, for example IgG-type antibodies.
  • FcyRIIb an Fc region
  • it is possible to engineer the Fc moiety to enhance FcyRIIB binding by introducing the mutations S267E and L328F as described by Chu, S. Y. et al., 2008: Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies.
  • Molecular Immunology 45, 3926-3933 are examples of the FcyRIIb.
  • the antibodies of the present disclosure comprise an engineered Fc moiety with the mutations S267E and L328F, in particular as described by Chu, S. Y. et al., 2008: Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies.
  • FcyRIIB may function to suppress further immunoglobulin production and isotype switching to, for example, the IgE class.
  • FcyRIIB On macrophages, FcyRIIB is thought to inhibit phagocytosis as mediated through FcyRIIA.
  • the B form On eosinophils and mast cells, the B form may help to suppress activation of these cells through IgE binding to its separate receptor.
  • modification in native IgG of at least one of E233- G236, P238, D265, N297, A327 and P329 reduces binding to FcyRI.
  • IgG2 residues at positions 233-236, substituted into corresponding positions IgGl and IgG4, reduces binding of IgGl and IgG4 to FcyRI by 10 3 -fold and eliminated the human monocyte response to antibody-sensitized red blood cells (Armour, K. L., et al. Eur J. Immunol. 29 (1999) 2613-2624).
  • FcyRIIA reduced binding for FcyRIIA is found, e.g., for IgG mutation of at least one of E233-G236, P238, D265, N297, A327, P329, D270, Q295, A327, R292 and K414.
  • FcyRIII binding reduced binding to FcyRIIIA is found, e.g., for mutation of at least one of E233-G236, P238, D265, N297, A327, P329, D270, Q295, Ki l, S239, E269, E293, Y296, V303, A327, K338 and D376. Mapping of the binding sites on human IgGl for Fc receptors, the above-mentioned mutation sites, and methods for measuring binding to FcyRI and FcyRIIA, are described in Shields, R. L., et al., J. Biol. Chem. 276 (2001) 6591-6604.
  • FcyRIIIA Two allelic forms of human FcyRIIIA are the "Fl 58" variant, which binds to IgGl Fc with lower affinity, and the "VI 58" variant, which binds to IgGl Fc with higher affinity. See, e.g., Bruhns et al., Blood 773:3716-3725 (2009).
  • two regions of native IgG Fc appear to be involved in interactions between FcyRIIs and IgGs, namely (i) the lower hinge site of IgG Fc, in particular amino acid residues L, L, G, G (234 - 237, EU numbering), and (ii) the adjacent region of the CH2 domain of IgG Fc, in particular a loop and strands in the upper CH2 domain adjacent to the lower hinge region, e.g. in a region of P331 (Wines, B.D., et al., J. Immunol. 2000; 164: 5313 - 5318).
  • FcyRI appears to bind to the same site on IgG Fc
  • FcRn and Protein A bind to a different site on IgG Fc, which appears to be at the CH2-CH3 interface
  • mutations that increase binding affinity of an Fc polypeptide or fragment thereof of the present disclosure to a (i.e., one or more) Fey receptor (e.g., as compared to a reference Fc polypeptide or fragment thereof or containing the same that does not comprise the mutation(s)). See, e.g., Delillo and Ravetch, Cell 161(5): 1035-1045 (2015) and Ahmed et al., J. Struc. Biol. 194(1):78 (2016), the Fc mutations and techniques of which are incorporated herein by reference.
  • an antibody or antigen-binding fragment can comprise a Fc polypeptide or fragment thereof comprising a mutation selected from G236A; S239D; A330L; and I332E; or a combination comprising any two or more of the same; e.g., S239D/I332E; S239D/A330L/I332E; G236A/S239D/I332E; G236A/A330L/I332E (also referred to herein as "GAALIE"); or G236A/S239D/A330L/I332E.
  • the Fc polypeptide or fragment thereof does not comprise S239D.
  • the Fc polypeptide or fragment thereof comprises S at position 239 (EU numbering). In certain embodiments, the Fc polypeptide or fragment thereof may comprise or consist of at least a portion of an Fc polypeptide or fragment thereof that is involved in FcRn binding.
  • the Fc polypeptide or fragment thereof comprises one or more amino acid modifications that improve binding affinity for (e.g., enhance binding to) FcRn (e.g., at a pH of about 6.0) and, in some embodiments, thereby extend in vivo half-life of a molecule comprising the Fc polypeptide or fragment thereof (e.g., as compared to a reference Fc polypeptide or fragment thereof or antibody that is otherwise the same but does not comprise the modification(s)).
  • FcRn e.g., at a pH of about 6.0
  • the Fc polypeptide or fragment thereof comprises or is derived from a IgG Fc and a half-life-extending mutation comprises any one or more of: M428L; N434S; N434H; N434A; N434S; M252Y; S254T; T256E; T250Q; P257I Q311I; D376V; T307A; E380A (EU numbering).
  • a half-life- extending mutation comprises M428L/N434S (also referred to herein as "MLNS", "LS", “_LS”, and "-LS”).
  • a half-life-extending mutation comprises M252Y/S254T/T256E.
  • a half-life-extending mutation comprises T250Q/M428L. In certain embodiments, a half-life-extending mutation comprises P257I/Q311I. In certain embodiments, a half-life-extending mutation comprises P257I/N434H. In certain embodiments, a half-life-extending mutation comprises D376V/N434H. In certain embodiments, a half-life-extending mutation comprises T307A/E380A/N434A. In certain embodiments, a half-life- extending mutation comprises M428L/N434A.
  • an antibody or antigen-binding fragment includes a Fc moiety that comprises the substitution mtuations M428L/N434S or M428L/N434A. In some embodiments, an antibody or antigen-binding fragment includes a Fc polypeptide or fragment thereof that comprises the substitution mtuations G236A/A330L/I332E. In certain embodiments, an antibody or antigen-binding fragment includes a (e.g., IgG) Fc moiety that comprises a G236A mutation, an A330L mutation, and a I332E mutation (GAALIE), and does not comprise a S239D mutation (e.g., comprises a native S at position 239).
  • a S239D mutation e.g., comprises a native S at position 239
  • an antibody or antigen-binding fragment includes an Fc polypeptide or fragment thereof that comprises the substitution mutations: M428L/N434S (or M428L/N434A) and G236A/A330L/I332E, and optionally does not comprise S239D (e.g., comprises S at 239).
  • an antibody or antigen-binding fragment includes a Fc polypeptide or fragment thereof that comprises the substitution mutations: M428L/N434S (or M428L/N434A) and G236A/S239D/A330L/I332E.
  • the antibody or antigen-binding fragment comprises a mutation that alters glycosylation, wherein the mutation that alters glycosylation comprises N297A, N297Q, or N297G, and/or the antibody or antigen-binding fragment is partially or fully aglycosylated and/or is partially or fully afucosylated.
  • Host cell lines and methods of making partially or fully aglycosylated or partially or fully afucosylated antibodies and antigen-binding fragments are known (see, e.g., PCT Publication No. WO 2016/181357; Suzuki et al. Clin. Cancer Res. 73(6):1875-82 (2007); Huang et al. MAbs 6 1-12 (2018)).
  • An antibody or antigen-binding fragment of the present disclosure can be fucosylated (e.g., comprising one or more fucosyl moiety, and typically comprising a native (wild-type) fucosylation pattern or a fucosylation pattern that includes one or more additional, or fewer, fucosyl moieties as compared to native), or can be afucosylated.
  • native IgGl antibodies carry a glycan site at N297, and this is typically the only site where a core fucose moiety may be found in the antibody, though some glycan sites may arise through mutation (e.g. in the variable domains) during antibody development.
  • Fucosylation of an Fc polypeptide or fragment thereof, or of an antibody can be effected by introducing amino acid mutations to introduce or disrupt a fucosylation site (e.g. a mutation at N297, such as N297Q or N297A, to disrupt formation of a glycan that can include a core fucose moiety), though typically it is preferred to maintain N297 and the glycan thereof, such as by expressing the polypeptide in a host cell which has been genetically engineered to lack the ability (or have an inhibited or compromised ability) to fucosylate the polypeptide; by expressing the polypeptide under conditions in which a host cell is impaired in its ability to fucosylate the polypeptide (e.g., in the presence of 2-fluoro-L-fucose (2FF)), or the like.
  • a fucosylation site e.g. a mutation at N297, such as N297Q or N297A, to disrupt formation of a glycan that can
  • An afucosylated polypeptide can comprise no fucose moieties, or substantially no fucose moieties, and/or can be expressed by a host cell that is genetically engineered to lack the ability (or have an inhibited or compromised ability) to fucosylate the polypeptide and/or can be expressed under conditions in which a host cell is impaired in its ability to fucosylate the polypeptide (e.g., in the presence of 2-fluoro-L-fucose (2FF)).
  • a polypeptide does not comprise a core fucose moiety at Asn297.
  • afucosylated polypeptides have increased binding to FcyRIIIA.
  • a plurality of antibodies may be described as “afucosylated” when the plurality was produced in the presence of 2FF or like reagent.
  • a plurality of polypeptides or antibodies may be described as, for example, afucosylated, meaning that about 85% or more of the single polypeptide or antibody molecules of the plurality do not comprise a fucose moiety.
  • an afucosylated antibody or polypeptide or a population or a plurality thereof comprises an asparagine (N) at EU position 297.
  • compositions are provided that comprise a plurality of any one or more of the presently disclosed polypeptides, wherein the composition comprises afucosylated polypeptides.
  • the antibody or antigen-binding fragment is capable of eliciting continued protection in vivo in a subject even once no detectable levels of the antibody or antigen-binding fragment can be found in the subject (z.e., when the antibody or antigen-binding fragment has been cleared from the subject following administration).
  • Such protection is referred to herein as a vaccinal effect.
  • dendritic cells can internalize complexes of antibody and antigen and thereafter induce or contribute to an endogenous immune response against antigen.
  • an antibody or antigenbinding fragment comprises one or more modifications, such as, for example, mutations in the Fc comprising G236A, A330L, and I332E, that are capable of activating dendritic cells that may induce, e.g., T cell immunity to the antigen.
  • an antibody or antigen-binding fragment fo the present disclosure comprises an Fc variant selected from the Fc variants summarized in Table C (see also PCT Publication No. WO 2022/251119).
  • the Fc variant, or the antibody or antigen-binding fragment is fucosylated.
  • the Fc variant, or the antibody or antigen-binding fragment is afucosylated.
  • an anti-parvovirus antibody or antigen-binding fragment comprises, in a(n e.g. human) IgGl heavy chain, the amino acid mutation(s) set forth in any one of (i)-(xviii): (i) G236A, L328V, and Q295E; (ii) G236A, P230A, and Q295E; (iii) G236A, R292P, and I377N; (iv) G236A, K334A, and Q295E; (v) G236S, R292P, and Y300L; (vi) G236A and Y300L; (vii) G236A, R292P, and Y300L; (viii) G236S, G420V, G446E, and L309T; (ix) G236A and R292P; (x) R292P and Y300L; (xi) G236A and R
  • the antibody or antigen-binding fragment is afucosylated.
  • the antibody or antigen-binding fragment further comprises one or more mutation that enhances binding to a human FcRn, such as M428L and N434S mutations or M428L and N434A mutations (EU numbering) or any other mutation(s) that enhance binding to a human FcRn, such as those described herein.
  • the antibody or antigen-binding fragment is afucosylated.
  • the antibody or antigen-binding fragment comprises a Fc polypeptide or a fragment thereof, including a CH2 (or a fragment thereof, a CH3 (or a fragment thereof), or a CH2 and a CH3, wherein the CH2, the CH3, or both can be of any isotype and may contain amino acid substitutions or other modifications as compared to a corresponding wild-type CH2 or CH3, respectively.
  • a Fc of the present disclosure comprises two CH2-CH3 polypeptides that associate to form a dimer.
  • the antibody or antigen-binding fragment can be monoclonal.
  • mAb monoclonal antibody
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, z.e., individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present, in some cases in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • polyclonal antibody preparations that include different antibodies directed against different epitopes, each monoclonal antibody is directed against a single epitope of the antigen.
  • the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
  • the term "monoclonal” is not to be construed as requiring production of the antibody by any particular method.
  • monoclonal antibodies useful in the present invention may be prepared by the hybridoma methodology first described by Kohler et al., Nature 256 :495 (1975), or may be made using recombinant DNA methods in bacterial, eukaryotic animal, or plant cells (see, e.g., U.S. Pat. No. 4,816,567). Monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example. Monoclonal antibodies may also be obtained using methods disclosed in PCT Publication No. WO 2004/076677A2.
  • Antibodies and antigen-binding fragments of the present disclosure include "chimeric antibodies" in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (see, U.S. Pat. Nos. 4,816,567; 5,530,101 and 7,498,415; and Morrison et al., Proc. Natl. Acad. Sci. USA, 57:6851-6855 (1984)).
  • chimeric antibodies may comprise human and non-human residues.
  • chimeric antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. For further details, see Jones et al.. Nature 321 :522-525 (1986); Riechmann et al., Nature 332:323- 329 (1988); and Presta, Curr. Op. Struct. Biol. 2:593-596 (1992). Chimeric antibodies also include primatized and humanized antibodies.
  • a “humanized antibody” is generally considered to be a human antibody that has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are typically taken from a variable domain. Humanization may be performed following the method of Winter and co-workers (Jones et al., Nature, 321 :522-525 (1986); Reichmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239: 1534-1536 (1988)), by substituting non-human variable sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized” antibodies are chimeric antibodies (U.S. Pat. Nos.
  • a "humanized" antibody is one which is produced by a non-human cell or animal and comprises human sequences, e.g., He domains.
  • human antibody is an antibody containing only sequences that are present in an antibody that is produced by a human (i.e., sequences that are encoded by human antibody-encoding genes).
  • human antibodies may comprise residues or modifications not found in a naturally occurring human antibody (e.g., an antibody that is isolated from a human), including those modifications and variant sequences described herein. These are typically made to further refine or enhance antibody performance.
  • human antibodies are produced by transgenic animals. For example, see U.S. Pat. Nos. 5,770,429; 6,596,541 and 7,049,426.
  • an antibody or antigen-binding fragment of the present disclosure is chimeric, humanized, or human.
  • PK pharmacokinetic
  • ti/2 or “half-life” refers to the elimination half-life of the antibody or antigen-binding fragment included in the pharmaceutical composition administered to a subject.
  • Clast generally refers to the last measurable plasma concentration (z.e., subsequent thereto, the substance is not present at a measurable concentration in plasma).
  • fusion proteins that comprise an antibody or antigen-binding fragment of the present disclosure.
  • a fusion protein comprises (i) an extracellular component comprising the antibody or antigen-binding fragment, (ii) a transmembrane component, and (iii) an intracellular component comprising one or more signaling domains (e.g., from CD3( ⁇ , CD28, 4-1BB, and/or TLR8).
  • a fusion protein comprises a chimeric antigen receptor.
  • a fusion protein comprises a chimeric engulfment receptor (see, e.g., Corey et al., Molecular Therapy Methods & Clinical Development 28: 1-10 (2023); doi.org/10.1016/j.omtim.2022.11.004).
  • the present disclosure provides isolated polynucleotides that encode any of the presently disclosed antibodies or an antigen-binding fragment thereof, or a portion thereof (e.g., a CDR, a VH, a VL, a heavy chain, or a light chain, or a heavy chain and a light chain), or that encode a presently disclosed polypeptide.
  • the polynucleotide comprises deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), wherein the RNA optionally comprises messenger RNA (mRNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • the polynucleotide comprises a modified nucleoside, a cap-1 structure, a cap-2 structure, or any combination thereof.
  • the polynucleotide comprises a pseudouridine, a N6-methyladenonsine, a 5- methylcytidine, a 2-thiouridine, or any combination thereof.
  • the pseudouridine comprises N1 -methylpseudouridine.
  • a polynucleotide comprises a polynucleotide having at least 50% (e.g., 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) identity to the polynucleotide sequence set forth in any one or more of SEQ ID NOs.: 6, 11, 16, 21, 26, and 31.
  • a polynucleotide comprises a polynucleotide having at least 50%, 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 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or more) identity to the polynucleotide sequence set forth in any one or more of SEQ ID NOs.: 6, 11, 16, 21, 26, and 31.
  • the polynucleotide is codon-optimized for expression in a host cell (e.g., a human cell, or a CHO cell).
  • a host cell e.g., a human cell, or a CHO cell.
  • codon optimization can be performed using known techniques and tools, e.g., using the GenScript® OptimiumGeneTM tool, or the like).
  • Codon-optimized sequences include sequences that are partially codon-optimized (z.e., one or more codon is optimized for expression in the host cell) and those that are fully codon-optimized.
  • a polynucleotide comprises the polynucleotide sequence of any one or more of SEQ ID NO.:6, 11, 16, 21, 26, and 31.
  • a polynucleotide comprises the polynucleotide sequence of SEQ ID NO. :6 and the polynucleotide sequence of SEQ ID NO. : 11.
  • a polynucleotide comprises the polynucleotide sequence of SEQ ID NO. : 16 and the polynucleotide sequence of SEQ ID NO. :21.
  • a polynucleotide comprises the polynucleotide sequence of SEQ ID NO. :26 and the polynucleotide sequence of SEQ ID NO. :31.
  • polynucleotides encoding antibodies and antigenbinding fragments of the present disclosure may possess different nucleotide sequences while still encoding a same antibody or antigen-binding fragment due to, for example, the degeneracy of the genetic code, splicing, and the like.
  • the polynucleotide can comprise deoxyribonucleic acid (DNA) or ribonucleic acid (RNA).
  • the RNA comprises messenger RNA (mRNA).
  • Vectors are also provided, wherein the vectors comprise or contain a polynucleotide as disclosed herein (e.g., a polynucleotide that encodes an anti- parvovirus antibody or antigen-binding fragment).
  • a vector can comprise any one or more of the vectors disclosed herein.
  • a vector is provided that comprises a DNA plasmid construct encoding the antibody or antigen-binding fragment, or a portion thereof (e.g., so-called "DMAb”; see, e.g., Muthumani et al., J Infect Dis.
  • a DNA plasmid construct comprises a single open reading frame encoding a heavy chain and a light chain (or a VH and a VL) of the antibody or antigenbinding fragment, wherein the sequence encoding the heavy chain and the sequence encoding the light chain are optionally separated by polynucleotide encoding a protease cleavage site and/or by a polynucleotide encoding a self-cleaving peptide.
  • the substituent components of the antibody or antigen-binding fragment are encoded by a polynucleotide comprised in a single plasmid.
  • the substituent components of the antibody or antigen-binding fragment are encoded by a polynucleotide comprised in two or more plasmids (e.g., a first plasmid comprises a polynucleotide encoding a heavy chain, VH, or VH+CH1, and a second plasmid comprises a polynucleotide encoding the cognate light chain, or VL).
  • a single plasmid comprises a polynucleotide encoding a heavy chain and/or a light chain from two or more antibodies or antigen-binding fragments of the present disclosure.
  • An exemplary expression vector is pVaxl, available from Invitrogen®.
  • a DNA plasmid of the present disclosure can be delivered to a subject by, for example, electroporation (e.g., intramuscular electroporation), or with an appropriate formulation (e.g., hyaluronidase).
  • a method comprises administering to a subject a first polynucleotide (e.g., mRNA) encoding an antibody heavy chain, a VH, or a Fd (VH + CHI), and administering to the subject a second polynucleotide (e.g., mRNA) encoding the cognate antibody light chain or VL.
  • a polynucleotide e.g., mRNA
  • a polynucleotide is provided that encodes a heavy chain and a light chain of an antibody or antigen-binding fragment thereof.
  • a polynucleotide e.g., mRNA
  • mRNA polynucleotide
  • a polynucleotide that encodes two heavy chains and two light chains of an antibody or antigen-binding fragment thereof. See, e.g. Li, JQ., Zhang, ZR., Zhang, HQ. et al. Intranasal delivery of replicating mRNA encoding neutralizing antibody against SARS-CoV-2 infection in mice. Sig Transduct Target Ther 6, 369 (2021). https://doi.org/10.1038/s41392-021-00783-l, the antibodyencoding mRNA constructs, vectors, and related techniques of which are incorporated herein by reference.
  • a polynucleotide is delivered to a subject via an alphavirus replicon particle (VRP) delivery system.
  • VRP alphavirus replicon particle
  • a replicon comprises a modified VEEV replicon comprising two subgenomic promoters.
  • a polynucleotide or replicon can translate simultaneously the heavy chain (or VH, or VH+1) and the light chain (or VL) of an antibody or antigenbinding fragment thereof.
  • a method is provided that comprises delivering to a subject such a polynucleotide or replicon.
  • the present disclosure also provides a host cell expressing an antibody or antigen-binding fragment according to the present disclosure; or comprising or containing a vector or polynucleotide according the present disclosure.
  • the cells include but are not limited to, eukaryotic cells, e.g., yeast cells, animal cells, insect cells, plant cells; and prokaryotic cells, including E. coli.
  • the cells are mammalian cells, such as human B cells.
  • the cells are a mammalian cell line such as CHO cells (e.g., DHFR- CHO cells (Urlaub et al., PNAS 77:4216 (1980)), human embryonic kidney cells (e.g., HEK293T cells), PER.C6 cells, Y0 cells, Sp2/0 cells.
  • NS0 cells human liver cells, e.g.
  • Hepa RG cells myeloma cells or hybridoma cells.
  • mammalian host cell lines include mouse sertoli cells (e.g., TM4 cells); monkey kidney CV1 line transformed by SV40 (COS-7); baby hamster kidney cells (BHK); African green monkey kidney cells (VERO-76); monkey kidney cells (CV1); human cervical carcinoma cells (HELA); human lung cells (W138); human liver cells (Hep G2); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3 A); mouse mammary tumor (MMT 060562); TRI cells; MRC 5 cells; and FS4 cells.
  • Mammalian host cell lines suitable for antibody production also include those described in, for example, Yazaki and
  • a host cell is a prokaryotic cell, such as an E. coli.
  • a prokaryotic cell such as an E. coli.
  • the expression of peptides in prokaryotic cells such as E. coli is well established (see, e.g., Pluckthun, A. Bio/Technology 9:545-551 (1991).
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • For expression of antibody fragments and polypeptides in bacteria see, e.g., U.S. Pat. Nos. 5,648,237; 5,789,199; and 5,840,523.
  • the cell may be transfected with a vector according to the present description with an expression vector.
  • transfection refers to the introduction of nucleic acid molecules, such as DNA or RNA (e.g., mRNA) molecules, into cells, such as into eukaryotic cells.
  • RNA e.g., mRNA
  • transfection encompasses any method known to the skilled person for introducing nucleic acid molecules into cells, such as into eukaryotic cells, including into mammalian cells.
  • Such methods encompass, for example, electroporation, lipofection, e.g., based on cationic lipids and/or liposomes, calcium phosphate precipitation, nanoparticle based transfection, virus based transfection, or transfection based on cationic polymers, such as DEAE-dextran or polyethylenimine, etc.
  • the introduction is non-viral.
  • host cells of the present disclosure may be transfected stably or transiently with a vector according to the present disclosure, e.g. for expressing an antibody, or an antigen-binding fragment thereof, according to the present disclosure.
  • the cells may be stably transfected with the vector as described herein.
  • cells may be transiently transfected with a vector according to the present disclosure encoding an antibody or antigen-binding fragment as disclosed herein.
  • a polynucleotide may be heterologous to the host cell. Accordingly, the present disclosure also provides recombinant host cells that heterologously express an antibody or antigen-binding fragment of the present disclosure.
  • the cell may be of a species that is different to the species from which the antibody was fully or partially obtained (e.g., CHO cells expressing a human antibody or an engineered human antibody).
  • the cell type of the host cell does not express the antibody or antigen-binding fragment in nature.
  • the host cell may impart a post-translational modification (PTM; e.g., glycosylation or fucosylation), or a lack thereof, on the antibody or antigenbinding fragment that is not present in a native state of the antibody or antigen-binding fragment (or in a native state of a parent antibody from which the antibody or antigen binding fragment was engineered or derived).
  • PTM post-translational modification
  • an antibody or antigen-binding fragment of the present disclosure that is produced by a host cell as disclosed herein may include one or more post-translational modification that is distinct from the antibody (or parent antibody) in its native state (e.g., a human antibody produced by a host cell can comprise one or more post-translational modification, or can include fewer post-translational modification(s), such that it is distinct from the antibody when isolated from the human and/or produced by the native human B cell or plasma cell).
  • Insect cells useful expressing a binding protein of the present disclosure include, for example, Spodoptera frugipera Sf9 cells, Trichoplusia ni BTI-TN5B1-4 cells, and Spodoptera frugipera SfSWTOl "MimicTM” cells. See, e.g., Palmberger et al., J. Biotechnol. 753(3-4): 160-166 (2011). Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Eukaryotic microbes such as filamentous fungi or yeast are also suitable hosts for cloning or expressing protein-encoding vectors, and include fungi and yeast strains with "humanized” glycosylation pathways, resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004); Li et al., Nat. Biotech. 24:210-215 (2006). Plant cells can also be utilized as hosts for expressing an antibody or antigenbinding fragment of the present disclosure.
  • PLANTIBODIESTM technology (described in, for example, U.S. Pat. Nos. 5,959,177; 6,040,498; 6,420,548; 7,125,978; and 6,417,429) employs transgenic plants to produce antibodies.
  • the host cell comprises a mammalian cell.
  • the host cell is a CHO cell, a HEK293 cell, a PER.C6 cell, a Y0 cell, a Sp2/0 cell, a NSO cell, a human liver cell, a myeloma cell, or a hybridoma cell.
  • the present disclosure provides methods for producing an antibody, or antigen-binding fragment, wherein the methods comprise culturing a host cell of the present disclosure under conditions and for a time sufficient to produce the antibody, or the antigen-binding fragment.
  • Methods useful for isolating and purifying recombinantly produced antibodies may include obtaining supernatants from suitable host cell/vector systems that secrete the recombinant antibody into culture media and then concentrating the media using a commercially available filter. Following concentration, the concentrate may be applied to a single suitable purification matrix or to a series of suitable matrices, such as an affinity matrix or an ion exchange resin.
  • One or more reverse phase HPLC steps may be employed to further purify a recombinant polypeptide. These purification methods may also be employed when isolating an immunogen from its natural environment. Methods for large scale production of one or more of the isolated/recombinant antibody described herein include batch cell culture, which is monitored and controlled to maintain appropriate culture conditions. Purification of soluble antibodies may be performed according to methods described herein and known in the art and that comport with laws and guidelines of domestic and foreign regulatory agencies.
  • compositions that comprise a presently disclosed antibody, antigen-binding fragment, polypeptide, polynucleotide, vector, or host cell, singly or in any combination, and can further comprise a pharmaceutically acceptable carrier, excipient, or diluent.
  • a pharmaceutically acceptable carrier excipient, or diluent.
  • a composition comprises a first vector comprising a first plasmid, and a second vector comprising a second plasmid, wherein the first plasmid comprises a polynucleotide encoding a heavy chain, VH, or VH+CH1, and a second plasmid comprises a polynucleotide encoding the cognate light chain or VL of the antibody or antigen-binding fragment thereof.
  • a composition comprises a polynucleotide (e.g., mRNA) coupled to a suitable delivery vehicle or carrier.
  • Exemplary vehicles or carriers for administration to a human subject include a lipid or lipid-derived delivery vehicle, such as a liposome, solid lipid nanoparticle, oily suspension, submicron lipid emulsion, lipid microbubble, inverse lipid micelle, cochlear liposome, lipid microtubule, lipid microcylinder, or lipid nanoparticle (LNP) or a nanoscale platform (see, e.g., Li et al. Wilery Interdiscip Rev. Nanomed Nanobiotechnol. 77(2):el530 (2019)).
  • LNP lipid nanoparticle
  • Principles, reagents, and techniques for designing appropriate mRNA and and formulating mRNA-LNP and delivering the same are described in, for example, Pardi et al.
  • lipid nanoparticles e.g., ionizable cationic lipid/phosphatidylcholine/cholesterol/PEG-lipid; ionizable lipid:distearoyl PC:cholesterol:polyethylene glycol lipid
  • subcutaneous, intramuscular, intradermal, intravenous, intraperitoneal, and intratracheal administration of the same, are incorporated herein by reference.
  • a composition comprises a first antibody or antigenbinding fragment of the present disclosure and a second antibody or antigen-binding fragment of the present disclosure, wherein of the first antibody or antigen-binding fragment and the second antibody or antigen-binding fragment are different.
  • the first antibody or antigen-binding fragment can be any anti-parvovirus antibody or antigenbinding fragment of the present disclosure
  • the second antibody or antigen-binding fragment can be any other anti-parvovirus antibody or antigen-binding fragment of the present disclosure.
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (see also Figure 8B)
  • the second antibody or antigenbinding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (see also Figure 8A).
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (see also Figure 8B)
  • the second antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18, as shown in Table A (see also Figure 8 A).
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • a parvovirus infection comprises infection with B 19.
  • a parvovirus infection comprises a chronic infection.
  • Methods of diagnosis may include contacting an antibody, antibody fragment (e.g., antigen binding fragment) with a sample.
  • samples may be isolated from a subject, for example an isolated tissue sample taken from, for example, nasal passages, sinus cavities, salivary glands, lung, liver, pancreas, kidney, ear, eye, placenta, alimentary tract, heart, ovaries, pituitary, adrenals, thyroid, brain, skin or blood.
  • the methods of diagnosis may also include the detection of an antigen/antibody complex, in particular following the contacting of an antibody or antibody fragment with a sample.
  • a detection step can be performed at the bench, i.e. without any contact to the human or animal body. Examples of detection methods are well-known to the person skilled in the art and include, e.g., ELISA (enzyme-linked immunosorbent assay), including direct, indirect, and sandwich ELISA.
  • a parvovirus infection comprises infection with Bl 9.
  • a parvovirus infection comprises a chronic parvovirus infection.
  • Treatment refers to medical management of a disease, disorder, or condition of a subject (e.g., a human or non-human mammal, such as a primate, horse, cat, dog, goat, mouse, or rat).
  • an appropriate dose or treatment regimen comprising an antibody or composition of the present disclosure is administered in an amount sufficient to elicit a therapeutic or prophylactic benefit.
  • Therapeutic or prophylactic/preventive benefit includes improved clinical outcome; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay or prevention of disease progression; remission; survival; prolonged survival; or any combination thereof.
  • therapeutic or prophylactic/preventive benefit includes reduction or prevention of hospitalization for treatment of a parvovirus infection (z.e., in a statistically significant manner).
  • therapeutic or prophylactic/preventive benefit includes a reduced duration of hospitalization for treatment of a parvovirus infection (z.e., in a statistically significant manner).
  • therapeutic or prophylactic/preventive benefit includes a reduced or abrogated need for respiratory intervention, such as intubation and/or the use of a respirator device.
  • therapeutic or prophylactic/preventive benefit includes reversing a late-stage disease pathology and/or reducing mortality.
  • a “therapeutically effective amount” or “effective amount” of an antibody, antigen-binding fragment, polynucleotide, vector, host cell, combination, or composition of this disclosure refers to an amount of the composition or molecule sufficient to result in a therapeutic effect, including improved clinical outcome; lessening or alleviation of symptoms associated with a disease; decreased occurrence of symptoms; improved quality of life; longer disease-free status; diminishment of extent of disease, stabilization of disease state; delay of disease progression; remission; survival; or prolonged survival in a statistically significant manner.
  • a therapeutically effective amount refers to the effects of that ingredient or cell expressing that ingredient alone.
  • a therapeutically effective amount refers to the combined amounts of active ingredients or combined adjunctive active ingredient with a cell expressing an active ingredient that results in a therapeutic effect, whether administered serially, sequentially, or simultaneously.
  • methods for treating a parvovirus infection in a subject, wherein the methods comprise administering to the subject an effective amount of an antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition as disclosed herein.
  • a method comprises administering to a subject an effective amount of: a first antibody or antigen-binding fragment of the present disclosure and a second antibody or antigen-binding fragment of the present disclosure (or administering an effective amount of a composition comprising the same), wherein of the first antibody or antigen-binding fragment and the second antibody or antigenbinding fragment are different.
  • the first antibody or antigen-binding fragment can be any anti-parvovirus antibody or antigen-binding fragment of the present disclosure
  • the second antibody or antigen-binding fragment can be any other anti-parvovirus antibody or antigen-binding fragment of the present disclosure.
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (see also Figure 8B)
  • the second antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (see also Figure 8A).
  • the first antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (see also Figure 8B)
  • the second antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigen-binding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18, as shown in Table A (see also Figure 8A).
  • the first antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (see also Figure 8B), and the second antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (see also Figure 8C).
  • the first antibody or antigen-binding fragment and/or the second antibody or antigenbinding fragment comprises a heavy chain comprising M428L and N434S mutations.
  • Subjects that can be treated by the present disclosure are, in general, human and other primate subjects, such as monkeys and apes for veterinary medicine purposes. Other model organisms, such as mice and rats, may also be treated according to the present disclosure.
  • the subject may be a human subject.
  • the subjects can be male or female and can be any suitable age, including infantjuvenile, adolescent, adult, and geriatric subjects.
  • a subject treated according to the present disclosure comprises one or more risk factors.
  • a human subject treated according to the present disclosure is an infant, a child, a young adult, an adult of middle age, or an elderly person. In certain embodiments, a human subject treated according to the present disclosure is less than 1 year old, or is 1 to 5 years old, or is between 5 and 125 years old (e.g., 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 125 years old, including any and all ages therein or therebetween).
  • a human subject treated according to the present disclosure is 0- 19 years old, 20-44 years old, 45-54 years old, 55-64 years old, 65-74 years old, 75-84 years old, or 85 years old, or older. Persons of middle, and especially of elderly age are can be at particular risk.
  • the human subject is 45-54 years old, 55-64 years old, 65-74 years old, 75-84 years old, or 85 years old, or older.
  • the human subject is male. In some embodiments, the human subject is female.
  • a subject receiving treatment for parvovirus infection has, or is at risk for having, contracting, or developing: fifth disease (z.e., a rash illness caused by parvovirus B19 and most commonly seen in children, also called erythema infetiosum); pure red cell aplasia; an immunocompromised state; sickle cell disease; rheumatoid arthritis; hydrops fetalis; fetal death; polyarthralgia; aplastic crisis (e.g., with increased hematopoiesis); or any combination of the foregoing.
  • fifth disease z.e., a rash illness caused by parvovirus B19 and most commonly seen in children, also called erythema infetiosum
  • pure red cell aplasia an immunocompromised state
  • sickle cell disease rheumatoid arthritis
  • hydrops fetalis fetal death
  • polyarthralgia aplastic crisis (e.g., with increased hematopoies
  • a subject receiving treatment for parvovirus infection according to the present disclosure is pregnant, has a chronic parvovirus infection, or both.
  • a subject receiving treatment for parvovirus infection according to the present disclosure has previously received an immune globulin preparation comprising immune globulin from a host or hosts (e.g., human and/or equine) that had been exposed to a parvovirus.
  • an immune globulin preparation comprising immune globulin from a host or hosts (e.g., human and/or equine) that had been exposed to a parvovirus.
  • Prophylaxis of infection with parvovirus refers in particular to prophylactic settings, wherein the subject was not diagnosed with infection with parvovirus (either no diagnosis was performed or diagnosis results were negative) and/or the subject does not show or experience symptoms of infection with parvovirus.
  • Prophylaxis of infection with parvovirus is particularly useful in subjects at greater risk of severe disease or complications when infected, such as pregnant women, children (such as children under 59 months), the elderly, individuals with chronic medical conditions (such as chronic cardiac, pulmonary, renal, metabolic, neurodevelopmental, liver or hematologic diseases) and individuals with immunosuppressive conditions (such as HIV/AIDS, receiving chemotherapy or steroids, or malignancy).
  • pregnant women such as children under 59 months
  • children such as children under 59 months
  • individuals with chronic medical conditions such as chronic cardiac, pulmonary, renal, metabolic, neurodevelopmental, liver or hematologic diseases
  • immunosuppressive conditions such as HIV/AIDS, receiving chemotherapy or steroids, or malignancy.
  • treatment is administered as peri-exposure or preexposure prophylaxis. In certain embodiments, treatment is administered as postexposure prophylaxis.
  • the subject is typically infected with parvovirus, diagnosed with parvovirus infection, and/or showing symptoms of parvovirus infection.
  • treatment and “therapy”/"therapeutic” of parvovirus infection can refer to (complete) cure as well as attenuation/reduction of parvovirus infection and/or related symptoms (e.g., attenuation/reduction of severity of infection and/or symptoms, number of symptoms, duration of infection and/or symptoms, or any combination thereof).
  • a reference subject can be, for example, (i) the same subject during an earlier period of time, (ii) a subject of a same or a similar: age or age group; gender; pregnancy status; chronic medical condition (such as chronic cardiac, pulmonary, renal, metabolic, neurodevel opmental, liver or hematologic diseases) or lack thereof; and/or immunosuppressive condition or lack thereof; or (iii) a typical subject within a population (e.g., local, regional, or national, including of a same or similar age or age range and/or general state of health).
  • Prophylaxis can be determined by, for example, the failure to develop a diagnosed parvovirus infection and/or the lack of symptoms associated with parvovirus infection.
  • treatment and/or prevention comprises post-exposure prophylaxis.
  • Typical routes of administering the presently disclosed compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques.
  • administering comprises administering by a route that is selected from oral, intravenous, parenteral, intragastric, intrapleural, intrapulmonary, intrarectal, intradermal, intraperitoneal, intratumoral, subcutaneous, topical, transdermal, intracisternal, intrathecal, intranasal, and intramuscular.
  • a method comprises orally administering the antibody, antigenbinding fragment, polynucleotide, vector, host cell, or composition to the subject.
  • compositions according to certain embodiments of the present invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a subject or patient may take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a herein described an antibody or antigen-binding in aerosol form may hold a plurality of dosage units.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000).
  • composition to be administered will, in any event, contain an effective amount of an antibody or antigen-binding fragment, polynucleotide, vector, host cell, , or composition of the present disclosure, for treatment of a disease or condition of interest in accordance with teachings herein.
  • a composition may be in the form of a solid or liquid.
  • the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form.
  • the carrier(s) may be liquid, with the compositions being, for example, an oral oil, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
  • the pharmaceutical composition is preferably in either solid or liquid form, where semi solid, semi liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
  • the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like.
  • a solid composition will typically contain one or more inert diluents or edible carriers.
  • binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
  • a liquid carrier such as polyethylene glycol or oil.
  • the composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred compositions contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • Liquid pharmaceutical compositions may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer’s solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Physiological saline is a preferred adjuvant.
  • a liquid composition intended for either parenteral or oral administration should contain an amount of an antibody or antigen-binding fragment as herein disclosed such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of the antibody or antigen-binding fragment in the composition. When intended for oral administration, this amount may be varied to be between 0.1 and about 70% of the weight of the composition. Certain oral pharmaceutical compositions contain between about 4% and about 75% of the antibody or antigen-binding fragment. In certain embodiments, pharmaceutical compositions and preparations according to the present invention are prepared so that a parenteral dosage unit contains between 0.01 to 10% by weight of antibody or antigen-binding fragment prior to dilution.
  • the composition may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base.
  • the base may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers. Thickening agents may be present in a composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device.
  • the pharmaceutical composition may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug.
  • the composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient.
  • bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
  • a composition may include various materials which modify the physical form of a solid or liquid dosage unit.
  • the composition may include materials that form a coating shell around the active ingredients.
  • the materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents.
  • the active ingredients may be encased in a gelatin capsule.
  • the composition in solid or liquid form may include an agent that binds to the antibody or antigen-binding fragment of the disclosure and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include monoclonal or polyclonal antibodies, one or more proteins or a liposome.
  • the composition may consist essentially of dosage units that can be administered as an aerosol.
  • aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols may be delivered in single phase, bi phasic, or tri phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One of ordinary skill in the art, without undue experimentation, may determine preferred aerosols.
  • compositions of the present disclosure also encompass carrier molecules for polynucleotides, as described herein (e.g., lipid nanoparticles, nanoscale delivery platforms, and the like).
  • compositions may be prepared by methodology well known in the pharmaceutical art.
  • a composition intended to be administered by injection can be prepared by combining a composition that comprises an antibody, antigen-binding fragment thereof, or antibody conjugate as described herein and optionally, one or more of salts, buffers and/or stabilizers, with sterile, distilled water so as to form a solution.
  • a surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalently interact with the peptide composition so as to facilitate dissolution or homogeneous suspension of the antibody or antigen-binding fragment thereof in the aqueous delivery system.
  • an appropriate dose and treatment regimen provide the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (such as described herein, including an improved clinical outcome (e.g., a decrease in frequency, duration, or severity of diarrhea or associated dehydration, or inflammation, or longer disease-free and/or overall survival, or a lessening of symptom severity).
  • a dose should be sufficient to prevent, delay the onset of, or diminish the severity of a disease associated with disease or disorder.
  • Prophylactic benefit of the compositions administered according to the methods described herein can be determined by performing pre-clinical (including in vitro and in vivo animal studies) and clinical studies and analyzing data obtained therefrom by appropriate statistical, biological, and clinical methods and techniques, all of which can readily be practiced by a person skilled in the art.
  • compositions are administered in an effective amount (e.g., to treat a parvovirus infection), which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the subject; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • an effective amount e.g., to treat a parvovirus infection
  • test subjects will exhibit about a 10% up to about a 99% reduction in one or more symptoms associated with the disease or disorder being treated as compared to placebo-treated or other suitable control subjects.
  • a therapeutically effective dose of an antibody or antigen binding fragment is (for a 70 kg mammal) from about 0.001 mg/kg (z.e., 0.07 mg) to about 100 mg/kg (z.e., 7.0 g); preferably a therapeutically effective dose is (for a 70 kg mammal) from about 0.01 mg/kg (z.e., 0.7 mg) to about 50 mg/kg (z.e., 3.5 g); more preferably a therapeutically effective dose is (for a 70 kg mammal) from about 1 mg/kg (z.e., 70 mg) to about 25 mg/kg (z.e., 1.75 g).
  • a therapeutically effective dose may be different than for an antibody or antigen-binding fragment.
  • a method comprises administering the antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition to the subject at 2, 3, 4, 5, 6, 7, 8, 9, 10 times, or more.
  • a method comprises administering the antibody, antigen-binding fragment, or composition to the subject a plurality of times, wherein a second or successive administration is performed at about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 24, about 48, about 74, about 96 hours, or more, following a first or prior administration, respectively.
  • a method comprises administering the antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition at least one time prior to the subject being infected by parvovirus.
  • compositions comprising an antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition of the present disclosure may also be administered simultaneously with, prior to, or after administration of one or more other therapeutic agents, such as, for example, human immunoglobulin preparations (IVIG).
  • IVIG human immunoglobulin preparations
  • Such combination therapy may include administration of a single pharmaceutical dosage formulation which contains a compound of the invention and one or more additional active agents, as well as administration of compositions comprising an antibody or antigen-binding fragment of the disclosure and each active agent in its own separate dosage formulation.
  • an antibody or antigen-binding fragment thereof as described herein and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations.
  • an antibody or antigenbinding fragment as described herein and the other active agent can be administered to the subject together in a single parenteral dosage composition such as in a saline solution or other physiologically acceptable solution, or each agent administered in separate parenteral dosage formulations.
  • a single parenteral dosage composition such as in a saline solution or other physiologically acceptable solution, or each agent administered in separate parenteral dosage formulations.
  • the compositions comprising an antibody or antigen-binding fragment and one or more additional active agents can be administered at essentially the same time, z.e., concurrently, or at separately staggered times, z.e., sequentially and in any order; combination therapy is understood to include all these regimens.
  • an antibody (or one or more nucleic acid, host cell, vector, or composition) is administered to a subject who has previously received one or more anti-inflammatory agent and/or one or more antiviral agent.
  • one or more anti-inflammatory agent and/or one or more antiviral agent is administered to a subject who has previously received an antibody (or one or more nucleic acid, host cell, vector, or composition).
  • uses of the presently disclosed antibodies, antigen-binding fragments, vectors, host cells, and compositions e.g., in the diagnosis, prophylaxis, and/or treatment of a parvovirus infection, in the manufacture of a medicament for preventing or treating a parvovirus infection.
  • an antibody, antigen-binding fragment, polynucleotide, vector, host cell, or composition is provided for use in a method of treating or preventing a parvovirus infection in a subject.
  • an antibody, antigen-binding fragment, or composition is provided for use in a method of manufacturing or preparing a medicament for treating or preventing a parvovirus infection in a subject.
  • the present disclosure also provides the following non-limiting enumerated Embodiments.
  • Embodiment 1 An anti -parvovirus antibody, or an antigen-binding fragment thereof.
  • Embodiment 2 The anti-parvovirus antibody or antigen-binding fragment of Embodiment 1, wherein the parvovirus comprises an erythrovirus, optionally wherein the parvovirus is an erythrovirus.
  • Embodiment 3 The anti-parvovirus antibody or antigen-binding fragment of Embodiment 1 or 2, wherein the parvovirus comprises Bl 9, optionally wherein the parvovirus is Bl 9.
  • Embodiment 4 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, which binds to an epitope in B19 uVPl.
  • Embodiment 5 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-4, which binds to a peptide having the amino acid sequence TGTDLE (SEQ ID NO.:4).
  • Embodiment 6 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-4, which binds to a peptide having the amino acid sequence X1X2TDX3EX4 (SEQ ID NO.:5), wherein:
  • Xi is present or absent, and, if present, is any amino acid
  • X2 is G or F, and is preferably G;
  • X3 is L or K, and is preferably K
  • X4 is present or absent, and, if present, is any amino acid except P.
  • Embodiment 7 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-6, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:22 wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • Embodiment 8 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-7, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • CDRL1 the CDRL2
  • CDRL3 the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:22
  • the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • Embodiment 9 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-7, comprising:
  • VH variable domain
  • CDR complementarity determining region
  • Embodiment 10 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-7 and 9, comprising:
  • the CDRL1 amino acid sequence set forth in SEQ ID NO.:23 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions
  • the CDRL2 amino acid sequence set forth in SEQ ID NO.:24 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • Embodiment 11 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-10, comprising the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.: 18-20 and 23-25, respectively.
  • Embodiment 12 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-11, comprising:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Embodiment 13 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-12, comprising:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Embodiment 14 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, which binds to an epitope in B19 VP2.
  • Embodiment 15 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • Embodiment 16 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 15, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • CDRL1 the CDRL2, and CDRL3 of the VL amino acid sequence set forth in SEQ ID NO. : 12, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • Embodiment 17 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14-15, comprising:
  • VH variable domain
  • CDR complementarity determining region
  • Embodiment 18 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, 15, and 17, comprising:
  • the CDRL1 amino acid sequence set forth in SEQ ID NO.: 13 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions
  • the CDRL2 amino acid sequence set forth in SEQ ID NO.: 14 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • Embodiment 19 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14-18, comprising the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.:8-10 and 13-15, respectively.
  • Embodiment 20 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14-19, comprising:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Embodiment 21 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14-20, comprising:
  • VH heavy chain variable domain
  • Embodiment 22 a light chain variable domain (VL) comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.: 12.
  • VL light chain variable domain
  • Embodiment 22 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3 and 14, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • CDRL1, the CDRL2, and/or the CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32 wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • Embodiment 23 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 22, comprising:
  • VH heavy chain variable domain
  • CDR complementarity determining region
  • VL light chain variable domain
  • CDRL1 the CDRL2, and CDRL3 of the VL amino acid sequence set forth in SEQ ID NO.:32, wherein the CDRs are defined in accordance with the IMGT, Kabat, Chothia, North, EU, Martin (Enhanced Chothia), Contact, AbM, CCG, or AHo numbering system, or in accordance with any combination thereof.
  • Embodiment 24 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 22, comprising:
  • VH variable domain
  • CDR complementarity determining region
  • Embodiment 25 in a/the light chain variable domain (VL), the complementarity determining region (CDR)L3 amino acid sequence set forth in SEQ ID NO.:35, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • Embodiment 25 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, 22, and 24, comprising:
  • CDR complementarity determining region
  • SEQ ID NO.:28 the complementarity determining region (CDR)H1 amino acid sequence set forth in SEQ ID NO.:28, or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions
  • CDR complementarity determining region
  • the CDRL1 amino acid sequence set forth in SEQ ID NO.:33 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions
  • the CDRL2 amino acid sequence set forth in SEQ ID NO.:34 or a functional variant thereof comprising one, two, or three, optionally conservative, amino acid substitutions.
  • Embodiment 26 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 22-25, comprising the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 amino acid sequences set forth in SEQ ID NOs.:28-30 and 33-35, respectively.
  • Embodiment 27 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 22-26 comprising:
  • VH heavy chain variable domain
  • a light chain variable domain comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to, or comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO.:32.
  • Embodiment 28. The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-3, 14, and 22-27, comprising:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Embodiment 29 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-28, wherein the anti -parvovirus antibody or antigenbinding fragment comprises a human antibody, a monoclonal antibody, a purified antibody, a single chain antibody, a Fab, a Fab’, a F(ab’)2, or Fv.
  • Embodiment 30 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-29, wherein the anti -parvovirus antibody or antigenbinding fragment is a multi-specific antibody or antigen-binding fragment, wherein, optionally, the antibody or antigen-binding fragment is a bi-specific antibody or antigen-binding fragment.
  • Embodiment 31 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-30, wherein the anti -parvovirus antibody or antigenbinding fragment comprises an (e.g., IgGl) Fc polypeptide or a fragment thereof.
  • an (e.g., IgGl) Fc polypeptide or a fragment thereof comprises an (e.g., IgGl) Fc polypeptide or a fragment thereof.
  • Embodiment 32 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-31, which comprises a IgG, IgA, IgM, IgE, or IgD isotype.
  • Embodiment 33 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-32, which comprises an IgG isotype selected from IgGl, IgG2, IgG3, and IgG4.
  • Embodiment 34 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-33, which comprises an IgGl isotype.
  • Embodiment 35 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-34, which comprises an IgGlm3 allotype, an IgGlml7 allotype, an IgGl ml allotype, or any combination thereof.
  • Embodiment 36 The anti-parvovirus antibody or antigen-binding fragment of Embodiment 31-35, wherein the Fc polypeptide or fragment thereof comprises:
  • a mutation that increases binding affinity to a human FcRn e.g., as measured using surface plasmon resonance (SPR) (e.g., Biacore, e.g., T200 instrument, using manufacturer’s protocols)
  • SPR surface plasmon resonance
  • Biacore e.g., T200 instrument, using manufacturer’s protocols
  • a mutation that increases binding affinity to a human FcyR e.g., as measured using surface plasmon resonance (SPR) (e.g., Biacore, e.g., T200 instrument, using manufacturer’s protocols, and/or as measured using mesoscale discovery (MSD))
  • SPR surface plasmon resonance
  • MSD mesoscale discovery
  • Embodiment 37 The anti-parvovirus antibody or antigen-binding fragment of Embodiment 36, wherein the mutation that increases binding affinity to a human FcRn comprises: M428L; N434S; N434H; N434A; N434S; M252Y; S254T; T256E; T250Q; P257I; Q311I; D376V; T307A; E380A; or any combination thereof.
  • Embodiment 38 The anti-parvovirus antibody or antigen-binding fragment of Embodiment 36 or 37, wherein the mutation that increases binding affinity to a human FcRn comprises: (i) M428L/N434S; (ii) M252Y/S254T/T256E; (iii) T250Q/M428L; (iv) P257VQ311I; (v) P257I/N434H; (vi) D376V/N434H; (vii) T307A/E380A/N434A; (viii) M428L/N434A; or (ix) any combination of (i)-(viii).
  • Embodiment 39 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 36-38, wherein the mutation that increases binding affinity to a human FcRn comprises M428L/N434S or M428L/N434A.
  • Embodiment 40 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 36-38, wherein the mutation that enhances binding to a FcyR comprises S239D; I332E; A330L; G236A; or any combination thereof.
  • Embodiment 41 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 36-40, wherein the mutation that enhances binding to a FcyR comprises: (i) S239D/I332E; (ii) S239D/A330L/I332E; (iii) G236A/S239D/I332E; or (iv) G236A/A330L/I332E, wherein the Fc polypeptide or fragment thereof optionally comprises Ser at position 239.
  • Embodiment 42 Embodiment 42.
  • Embodiment 43 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-42, which: is afucosylated; has been produced in a host cell that is incapable of fucosylation or that is inhibited in its ability to fucosylate a polypeptide; has been produced under conditions that inhibit fucosylation thereof by a host cell; or any combination thereof.
  • Embodiment 44 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-43, which is human, humanized, or chimeric.
  • Embodiment 45 An isolated polynucleotide encoding the anti -parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-44, or encoding a VH, a heavy chain, a VL, and/or a light chain of the anti-parvovirus antibody or the antigen-binding fragment.
  • Embodiment 46 The polynucleotide of Embodiment 45, wherein the polynucleotide comprises deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), wherein the RNA optionally comprises messenger RNA (mRNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • Embodiment 47 The polynucleotide of any one of Embodiments 45-46, comprising a modified nucleoside, a cap-1 structure, a cap-2 structure, or any combination thereof.
  • Embodiment 48 The polynucleotide of Embodiment 47, wherein the polynucleotide comprises a pseudouridine, a N6-methyladenonsine, a 5-methylcytidine, a 2-thiouridine, or any combination thereof.
  • Embodiment 49 The polynucleotide of Embodiment 48, wherein the pseudouridine comprises N1 -methylpseudouridine.
  • Embodiment 51 The polynucleotide of Embodiment 50, wherein the host cell comprises a human cell.
  • Embodiment 52 A recombinant vector comprising the polynucleotide of any one of Embodiments 45-51.
  • Embodiment 53 A host cell comprising the polynucleotide of any one of Embodiments 45-51 and/or the vector of Embodiment 52, wherein the polynucleotide is optionally heterologous to the host cell and/or wherein the host cell is capable of expressing the encoded antibody or antigen-binding fragment or polypeptide.
  • Embodiment 54 An isolated human B cell comprising the polynucleotide of any one of Embodiments 45-51 and/or the vector of Embodiment 52, wherein polynucleotide is optionally heterologous to the human B cell and/or wherein the human B cell is immortalized.
  • Embodiment 55 A composition comprising:
  • Embodiment 56 The composition of Embodiment 55, comprising a first anti-parvovirus antibody or antigen-binding fragment and a second anti-parvovirus antibody or antigen-binding fragment, wherein each of the first antibody or antigenbinding fragment and the second antibody or antigen-binding fragment are different and are each according any one of Embodiments 1-44, wherein, preferably, the first antiparvovirus antibody or antigen-binding fragment binds to an epitope in B19 uVPl and the second anti-parvovirus antibody or antigen-binding fragment binds to an epitope in B19 VP2, wherein, optionally:
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (z.e., comprises SEQ ID NOs.: 18-20 and 23-25), and the second antiparvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (z.e., comprises SEQ ID NOs.:8-10 and 13-15);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A
  • the second anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (z.e., comprises SEQ ID NOs.:28-30 and 33-35);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (z.e., comprises SEQ ID NOs.: 17 and 22), and the second anti -parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18 (z.e., comprises SEQ ID NOs.:7 and 12), as shown in Table A; or
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A
  • the second antiparvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (z.e., comprises SEQ ID NOs.:27 and 32).
  • Embodiment 57 A composition comprising the polynucleotide of any one of Embodiments 45-51 or the vector of Embodiment 52 encapsulated in a carrier molecule, wherein the carrier molecule optionally comprises a lipid, a lipid-derived delivery vehicle, such as a liposome, a solid lipid nanoparticle, an oily suspension, a submicron lipid emulsion, a lipid microbubble, an inverse lipid micelle, a cochlear liposome, a lipid microtubule, a lipid microcylinder, lipid nanoparticle (LNP), or a nanoscale platform.
  • a lipid-derived delivery vehicle such as a liposome, a solid lipid nanoparticle, an oily suspension, a submicron lipid emulsion, a lipid microbubble, an inverse lipid micelle, a cochlear liposome, a lipid microtubule, a lipid microcylinder, lipid nanop
  • Embodiment 58 A method of making an anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-44, comprising culturing the host cell of Embodiment 53 or the human B cell of Embodiment 54 for a time and under conditions sufficient for the host cell or human B cell, respectively, to express the anti-parvovirus antibody or antigen-binding fragment.
  • Embodiment 59 The method of Embodiment 58, further comprising isolating the anti-parvovirus antibody or antigen-binding fragment.
  • Embodiment 60 A method of treating or preventing a parvovirus infection in a subject, the method comprising administering to the subject an effective amount of:
  • composition of any one of Embodiments 55-57 wherein, optionally, the method comprises administering to the subject an effective amount: of a first antibody or antigen-binding fragment and a second antibody or antigen-binding fragment, wherein, further optionally:
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (z.e., comprises SEQ ID NOs.: 18-20 and 23-25), and the second antiparvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (i.e., comprises SEQ ID NOs.:8-10 and 13-15);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A
  • the second anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A i.e., comprises SEQ ID NOs.:28-30 and 33-35);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A i.e., comprises SEQ ID NOs.: 17 and 22
  • the second anti -parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18 i.e., comprises SEQ ID NOs.:7 and 12), as shown in Table A
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A
  • the second antiparvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (z.e., comprises SEQ ID NOs.:27 and 32).
  • Embodiment 61 The method of Embodiment 60, comprising administering a single dose of the anti-parvovirus antibody or antigen-binding fragment, polynucleotide, recombinant vector, host cell, or composition to the subject.
  • Embodiment 62 The method of Embodiment 60 or 61, comprising administering two or more doses of the anti-parvovirus antibody or antigen-binding fragment, polynucleotide, recombinant vector, host cell, or composition to the subject.
  • Embodiment 63 The method of any one of Embodiments 60-62, comprising administering the anti-parvovirus antibody or antigen-binding fragment, polynucleotide, recombinant vector, host cell, or composition intramuscularly, subcutaneously, or intravenously.
  • Embodiment 64 The method of any one of Embodiments 60-63, wherein the treatment and/or prevention comprises post-exposure prophylaxis.
  • Embodiment 65 The method of any one of Embodiments 60-64, wherein the subject has received, is receiving, or will receive an antiviral.
  • Embodiment 66 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-44, the polynucleotide of any one of Embodiments 45-51, the recombinant vector of Embodiment 52, the host cell of Embodiment 53, the human B cell of Embodiment 54, and/or the composition of any one of Embodiments 55-57, for use in a method of treating or preventing a parvovirus infection in a subject wherein, optionally, the method comprises administering to the subject an effective amount: of a first antibody or antigen-binding fragment and a second antibody or antigen-binding fragment, wherein, further optionally:
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (z.e., comprises SEQ ID NOs.: 18-20 and 23-25), and the second antiparvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (i.e., comprises SEQ ID NOs.:8-10 and 13-15);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A
  • the second anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (i.e., comprises SEQ ID NOs.:28-30 and 33-35);
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (i.e., comprises SEQ ID NOs.: 17 and 22), and the second anti -parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18 (i.e., comprises SEQ ID NOs.:7 and 12), as shown in Table A; or
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A
  • the second antiparvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (z.e., comprises SEQ ID NOs.:27 and 32).
  • Embodiment 67 The anti-parvovirus antibody or antigen-binding fragment of any one of Embodiments 1-44, the polynucleotide of any one of Embodiments 45-51, the recombinant vector of Embodiment 52, the host cell of Embodiment 53, the human B cell of Embodiment 54, and/or the composition of any one of Embodiments 55-57, for use in the preparation of a medicament for the treatment or prevention of a parvovirus infection in a subject, wherein, optionally, the medicament comprises an effective amount: of a first antibody or antigen-binding fragment and a second antibody or antigen-binding fragment, wherein, further optionally:
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A (i.e., comprises SEQ ID NOs.: 18-20 and 23-25), and the second antiparvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB18, as shown in Table A (i.e., comprises SEQ ID NOs.:8-10 and 13-15); (b) the first anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAB5, as shown in Table A, and the second anti-parvovirus antibody or antigen-binding fragment comprises the CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3 of antibody PAA19, as shown in Table A (i.e., comprises SEQ
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A (i.e., comprises SEQ ID NOs.: 17 and 22), and the second anti -parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB18 (i.e., comprises SEQ ID NOs.:7 and 12), as shown in Table A; or
  • the first anti-parvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAB5, as shown in Table A
  • the second antiparvovirus antibody or antigen-binding fragment comprises the VH and the VL of antibody PAA19, as shown in Table A (z.e., comprises SEQ ID NOs.:27 and 32).
  • Embodiment 68 A method for in vitro diagnosis of a parvovirus infection, the method comprising:
  • Embodiment 69 The method of any one of Embodiments 60-65 and 68 or the anti-parvovirus antibody or antigen-binding fragment, polynucleotide, recombinant vector, host cell, human B cell, and/or composition for use of Embodiment 66 or 67, wherein the parvovirus infection comprises a B19 infection and/or comprises a chronic parvovirus infection.
  • the parvovirus infection comprises a B19 infection and/or comprises a chronic parvovirus infection.
  • Supernatants from stimulated B cells from 30 healthy donor tonsils were screened for the presence of IgG binding to Parvovirus B19 VPl/VP2-expressing viruslike particles (VLPs) using an antigen-specific B-cell memory repertoire analysis (AMBRA) method ( Figure 1). Half of the donors are positive to VP1/VP2. Forty-three percent (43%) of the tonsil donors scored positive. Three tonsils were selected for isolation of mAbs.
  • IgG + memory B cells from the three selected cryopreserved tonsils were immortalized with EBV and TLR agonist as described (Traggiai et al., Nat Med 2004, the techniques of which are incorporated herein by reference), and interrogated for the presence of antibodies against VLPs displaying VP1/VP2 or VP2 alone (native conformation ofVPl and VP2).
  • Figure 2A shows frequency of positivity from AMBRA supernatants on VP1/VP2 and VP2 VLPs.
  • Figures 2B-2C show endpoint VP 1 vs VP2 from 60 Sera collected in Bologna, Italy ( Figure 2B) or 111 sera collected in Bellinzona, Switzerland ( Figure 2C): Ab values were expressed as endpoint titers defined as the highest dilution at which the OD was higher than twice the background OD.
  • Figure 2D shows inhibition of competition in sera collected in Bellinzona with PAB5 (VP1) biotinylated vs PAB18 (VP2) biotinylated.
  • Figure 3A provides a table showing, for the indicated mAbs (PAB5-PAC69), neutralization of mAb, the light chain isotype of mAb determined by ELISA, the germline usage for V-D-J (VH) and V-J (VL) genes defined using the ImMunoGeneTics information system (IMGT) database, and results of Western blot binding to reduced and non-reduced antigen.
  • PAB5-PAC69 the indicated mAbs
  • VH V-D-J
  • VL V-J
  • Binding of various mAbs isolated from immortalized IgG+ memory B cells was assessed by ELISA ( Figures 3B-3D). Binding to VP1/VP2 VLPs or VP2-only VLPs was used to determine the specificity of isolated mAbs so that VP 1 -specific mAbs bound to VP1/VP2 VLPs only, while VP2-specific mAbs bound to both VP1/VP2 VLPs and VP2 VLPs. Six anti-VPl and nine anti-VP2 mAbs were selected from the interrogation of memory IgG B cells derived from selected tonsils. Six mAbs were found to bind specifically to VP1 on VP1/VP2 VLPs.
  • PBS-T PBS 0.05% Tween 20
  • pNPP 4-nitrophenyl phosphate
  • Figure 3C shows mAbs binding to VP1/VP2 and VP2 VLPs (i.e., VP2-specific) (PAC57, PAC58, PAC59 are not recombinant).
  • Figure 3D shows IgG and Fab of PAB5 mAb binding to VP1/VP2 VLPs.
  • Binding and blockade of binding (BOB) for IVIG (Grifols Pharmaceuticals), PAB5 and PAB18 mabs to VP1/VP2 VLPs were also compared.
  • Figure 4A shows binding of IVIG, PAB5 and PAB18 mAbs to uVPl/VP2 VLPs by ELISA.
  • Blockade of binding of labeled PAB5 ( Figure 4B) or PAB18 ( Figure 4C) mAbs to VP1/VP2 VLPs by different concentrations of unlabeled PAB5, PAB18, and IVIG was assessed.
  • Identified anti- VP 1 and anti-VP2 IgGs were tested in neutralization assays using seronegative patient sera from primary B19 infections containing B 19 on UT7/Epo-Sl cells. After 3 days, the level of neutralization was measured by immunofluorescence analysis after permeabilization using a 488DL-anti-B19. Data on EPC primary cells showed potent neutralization by the selected VP1 and VP2 antibodies ( Figures 6A-6C). Fc-mediated neutralization was exhibited by PAA19 and partially by PAB5, but not by PAB18.
  • VP1-I to VP1-IV Four uVPl antigenic sites (VP1-I to VP1-IV) were defined using the 6 VP1- specific mAbs: PAB5 and PAC21 were assigned to the VP1-I site, PAB47, PAC4 and PAC10 to VP1-II, VP1-III and VP1-IV, respectively ( Figure 5A). Two VP2 antigenic sites were defined with the 9 VP2-specific mAbs tested. PAB18, PAA21, PAA23 and PAC60 mAbs were assigned to the VP2-I antigenic site, while PAA19 was assigned to a unique VP2-II antigenic site due to the lack of competition with VP2-I mAbs such as PAB18 and PAC60.
  • PAC40, PAC58 and PAC57 were assigned to a site overlapping VP2-I and VP2-II (VP2-I/II).
  • PAC69 mAb was not assigned to a specific site due to the observed pattern of competition with PAC40 and PAC58 mAbs but not with the other VP1-I site mAbs ( Figure 5B).
  • MAbs were labeled with biotin and tested by ELISA in a matrix competition assay, in which unlabeled antibodies were incubated first at a concentration of 5 pg/ml on VP2- or VP1/VP2 VLPs coated plates (coating in PBS at 2 pg/ml), followed by the addition of a limiting concentration of biotinylated antibodies whose binding was revealed with alkaline phosphatase-conjugated streptavidin.
  • competition results it should be taken into account that if two epitopes overlap, or the areas covered by the arms of the two antibodies overlap, competition should be almost complete. Weak inhibitory or enhancing effects may reflect a decrease in affinity owing to steric or allosteric effects.
  • FIG. 5C shows an intensity plot with averaged spot intensities of PAB5-rIgGl (lOpg/ml) on a conformational peptide microarray containing 696 different cyclic constrained peptides.
  • Figure 5D shows an intensity plot with averaged spot intensities of PAA19-rIgGl (500 pg/ml) on a conformational peptide microarray containing 1,677 different cyclic constrained peptides.
  • the intensity plots were correlated with peptide and intensity maps as well as with visual inspection of the microarray scans to identify the epitopes of the human antibodies. In case it was not clear if a certain amino acid contributed to antibody binding, the corresponding letter was written in lighter font.
  • Figure 5E shows an amino acid plot of PAB5-rIgGl against the microarray containing conformational peptides with 119 variants of the wild type peptide: the plot was calculated by dividing the spot intensity of a given peptide (e.g., 1 YPYDVQDYA 9 ) (SEQ ID NO.:44) by the spot intensity of the native epitope ( 1 YPYDVPDYA 9 ) (SEQ ID NO.:45). The position of an amino acid at a given position, thus, reflects the intensity ratio compared to the wild type amino acid at the same position.
  • the heat map highlights a conserved core motif 2 GTDLE 6 (SEQ ID NO.:46).
  • Amino acid positions 3 T, 4 D and 6 E were essential for binding by PAB5, as all amino acid changes were not tolerated. Positions 2 G and 5 L display only a very limited tolerance for substitutions. The N- and C-terminal amino acid positions ’T and 7 L are less essential for binding.
  • the VP1 RBD (5-80 aa) includes a rigid fold of three alpha helices (with conserved aa).
  • the peptide TGTDLE (SEQ ID NO. :4) is in the loop region between helix 1 and helix 2. Mutation on TGTDLE (SEQ ID NO.:4) changes the spatial assembly of the three helices. Analysis of all permutations of the epitope was performed ( Figure 7).
  • FIG. 9A shows neutralization obtained from an experiment performed with titration of the antibodies or combo.
  • Figure 9B shows neutralization obtained from an experiment with further titrations of the antibodies or combo.
  • Figure 9C shows neutralization obtained from an experiment with 1 ug/ml of antibody or combo.
  • Neutralization of infection was also assessed via nucleic acid analysis. Briefly, equal amounts of 1 xlO 5 cells were collected at 2 and 72 hpi following infection and processed by using the Maxwell Viral Total Nucleic Acid kit on a Maxwell MDx platform (Promega), to obtain a total nucleic acid fraction in elution volumes of 100 pL. For quantitative analysis of viral DNA, an aliquot of the eluted nucleic acids, corresponding to 500 cells, was amplified by qPCR (Maxima SYBR Green qPCR Master Mix, Thermo Scientific) in a RotorQ system (Qiagen).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente divulgation concerne des anticorps et des fragments de liaison à l'antigène de ceux-ci qui peuvent se lier au parvovirus, par exemple au parvovirus B19, et peuvent neutraliser une infection à parvovirus.<i /> L'invention concerne également des polynucléotides qui codent pour un anticorps, des vecteurs qui comprennent de tels polynucléotides, des cellules hôtes qui peuvent exprimer les anticorps, des compositions associées, ainsi que des méthodes d'utilisation des compositions divulguées pour, par exemple, traiter ou prévenir une infection à parvovirus.
PCT/US2023/068460 2022-06-15 2023-06-14 Anticorps anti-parvovirus et leurs utilisations WO2023245078A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263352600P 2022-06-15 2022-06-15
US63/352,600 2022-06-15

Publications (1)

Publication Number Publication Date
WO2023245078A1 true WO2023245078A1 (fr) 2023-12-21

Family

ID=87280751

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/068460 WO2023245078A1 (fr) 2022-06-15 2023-06-14 Anticorps anti-parvovirus et leurs utilisations

Country Status (1)

Country Link
WO (1) WO2023245078A1 (fr)

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5283173A (en) 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
WO2004076677A2 (fr) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Production d'anticorps monoclonaux par transformation de lymphocytes b par le virus d'epstein barr
US7049426B2 (en) 1999-06-10 2006-05-23 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
US7498415B2 (en) 2003-09-24 2009-03-03 Kyowa Hakko Kogyo Co., Ltd. Recombinant antibody against human insulin-like growth factor
US8119772B2 (en) 2006-09-29 2012-02-21 California Institute Of Technology MART-1 T cell receptors
US8258268B2 (en) 2005-08-19 2012-09-04 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
WO2015103072A1 (fr) 2013-12-30 2015-07-09 Epimab Biotherapeutics Fabs d'immunoglobuline en tandem et leurs utilisations
WO2016181357A1 (fr) 2015-05-13 2016-11-17 Zumutor Biologics, Inc. Protéine afucosylée, cellule exprimant ladite protéine et procédés associés
WO2019025391A1 (fr) 2017-07-31 2019-02-07 Institute For Research In Biomedicine Anticorps ayant des domaines fonctionnels dans la région de coude entre un domaine variable et un domaine constant
WO2019057122A1 (fr) 2017-09-22 2019-03-28 Wuxi Biologics (Shanghai) Co., Ltd. Nouveaux complexes polypeptidiques bispécifiques
WO2022251119A2 (fr) 2021-05-24 2022-12-01 Vir Biotechnology, Inc. Polypeptides modifiés

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5283173A (en) 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5468614A (en) 1990-01-24 1995-11-21 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US7049426B2 (en) 1999-06-10 2006-05-23 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
WO2004076677A2 (fr) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Production d'anticorps monoclonaux par transformation de lymphocytes b par le virus d'epstein barr
US7498415B2 (en) 2003-09-24 2009-03-03 Kyowa Hakko Kogyo Co., Ltd. Recombinant antibody against human insulin-like growth factor
US8258268B2 (en) 2005-08-19 2012-09-04 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US8119772B2 (en) 2006-09-29 2012-02-21 California Institute Of Technology MART-1 T cell receptors
WO2015103072A1 (fr) 2013-12-30 2015-07-09 Epimab Biotherapeutics Fabs d'immunoglobuline en tandem et leurs utilisations
WO2016181357A1 (fr) 2015-05-13 2016-11-17 Zumutor Biologics, Inc. Protéine afucosylée, cellule exprimant ladite protéine et procédés associés
WO2019025391A1 (fr) 2017-07-31 2019-02-07 Institute For Research In Biomedicine Anticorps ayant des domaines fonctionnels dans la région de coude entre un domaine variable et un domaine constant
WO2019024979A1 (fr) 2017-07-31 2019-02-07 Institute For Research In Biomedicine Anticorps à domaines fonctionnels dans la région de coude
WO2019057122A1 (fr) 2017-09-22 2019-03-28 Wuxi Biologics (Shanghai) Co., Ltd. Nouveaux complexes polypeptidiques bispécifiques
WO2022251119A2 (fr) 2021-05-24 2022-12-01 Vir Biotechnology, Inc. Polypeptides modifiés

Non-Patent Citations (86)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2000, PHILADELPHIA COLLEGE OF PHARMACY AND SCIENCE
ABHINANDANMARTIN, MOL IMMUNOL., vol. 45, 2008, pages 3832 - 9
AHMED ET AL., J. STRUC. BIOL., vol. 194, no. 1, 2016, pages 78
ALTSCHUL ET AL., NUCLEIC ACIDS RES, vol. 25, 1997, pages 3389 - 3402
ANDERSON S ET AL: "Peptides derived from the unique region of B19 parvovirus minor capsid protein elicitneutralizing antibodies in rabbits", VIROLOGY, ELSEVIER, AMSTERDAM, NL, vol. 206, no. 1, 10 January 1995 (1995-01-10), pages 626 - 632, XP004786686, ISSN: 0042-6822, DOI: 10.1016/S0042-6822(95)80079-4 *
ARMOUR, K. L. ET AL., EUR. J. IMMUNOL., vol. 29, 1999, pages 2613 - 2624
BILKOVA ET AL., VIRUSES, vol. 6, no. 7, 2014, pages 2899 - 2937
BIOINFORMATICS, vol. 15, 2016, pages 298 - 300
BRINKMANNKONTERMANN, MABS, vol. 9, no. 2, 2017, pages 182 - 212
BRUHNS ET AL., BLOOD, vol. 113, 2009, pages 3716 - 3725
BURTON, D. R., MOL. IMMUNOL., vol. 22, 1985, pages 161 - 206
CAPEL, P. J. ET AL., IMMUNOMETHODS, vol. 113, 1994, pages 269 - 315
CHAUDHARY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 1066 - 1070
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHU, S. ET AL.: "Accelerated Clearance of IgE In Chimpanzees Is Mediated By Xmab7195, An Fc-Engineered Antibody With Enhanced Affinity For Inhibitory Receptor FcyRIIb", AM J RESPIR CRIT, AMERICAN THORACIC SOCIETY INTERNATIONAL CONFERENCE ABSTRACTS, 2014
CHU, S. Y ET AL.: "Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies", MOLECULAR IMMUNOLOGY, vol. 45, 2008, pages 3926 - 3933, XP002498116, DOI: 10.1016/j.molimm.2008.06.027
CHU, S. Y ET AL.: "Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies", MOLECULAR, vol. 45, 2008, pages 3926 - 3933, XP002498116, DOI: 10.1016/j.molimm.2008.06.027
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
COFFIN, J. M. ET AL.: "Fundamental Virology", 1996, LIPPINCOTT-RAVEN PUBLISHERS, article "Retroviridae: The viruses and their replication"
COREY ET AL., MOLECULAR THERAPY METHODS & CLINICAL DEVELOPMENT, vol. 28, 2023, pages 1 - 10
DE HAAS, M. ET AL., J LAB. CLIN. MED., vol. 126, 1995, pages 330 - 341
DELILLORAVETCH, CELL, vol. 161, no. 5, 2015, pages 1035 - 1045
DORSCH SIMONE ET AL: "The VP1-unique region of parvovirus B19: amino acid variability and antigenic stability", JOURNAL OF GENERAL VIROLOGY, vol. 82, no. 1, 1 January 2001 (2001-01-01), pages 191 - 199, XP093078084, ISSN: 0022-1317, Retrieved from the Internet <URL:https://www.microbiologyresearch.org/docserver/fulltext/jgv/82/1/0820191a.pdf?expires=1693565312&id=id&accname=guest&checksum=F6F0595B573D0A0D1AE8E8230FD0D464> DOI: 10.1099/0022-1317-82-1-191 *
DRECHSLER M D ET AL: "Generation of multifunctional murine monoclonal antibodies specifically directed to the VP1unique region protein of human parvovirus B19", IMMUNOBIOLOGY, URBAN UND FISCHER VERLAG, DE, vol. 213, no. 6, 10 July 2008 (2008-07-10), pages 511 - 517, XP022695118, ISSN: 0171-2985, [retrieved on 20080129], DOI: 10.1016/J.IMBIO.2007.12.003 *
DUNCAN, A. R.WINTER, G., NATURE, vol. 332, 1988, pages 323 - 327
ELLIOTT ET AL., NPJ VACCINES, vol. 18, 2017
ENGELS ET AL., HUM. GENE THER., vol. 77, 2003, pages 1155
EYLER ET AL., PNAS, vol. 116, no. 46, 2019, pages 23068 - 23071
FLINGAI ET AL., SCI REP, vol. 5, 2015, pages 12616
FRECHA ET AL., MOL. THER., vol. 18, 2010, pages 1748
GANESAN, L. P. ET AL.: "FcyRIIb on liver sinusoidal endothelium clears small immune complexes", JOURNAL OF IMMUNOLOGY, vol. 189, 2012, pages 4981 - 4988, XP002724347, DOI: 10.4049/jimmunol.1202017
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GESSNER, J. E. ET AL., ANN. HEMATOL., vol. 76, 1998, pages 231 - 248
GEURTS ET AL., MOL. THER., vol. 8, 2003, pages 108
GIGLER ET AL: "Generation of neutralizing human monoclonal antibodies against parvovirus B19 proteins.", JOURNAL OF VIROLOGY, vol. 73, no. 3, 1 March 1999 (1999-03-01), pages 1974 - 1979, XP055026464, ISSN: 0022-538X *
HONEGGERPLUCKTHUN, J. MOL. BIO., vol. 309, 2001, pages 657 - 670
HUANG ET AL., MABS, vol. 6, 2018, pages 1 - 12
JOLLY, D J: "The Development of Human Gene Therapy", 1999, COLD SPRING HARBOR LAB, article "Emerging Viral Vectors", pages: 209 - 40
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KERR J R ET AL: "Design and production of a target-specific monoclonal antibody to parvovirus B19 capsid proteins", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 180, no. 1, 13 March 1995 (1995-03-13), pages 101 - 106, XP004021075, ISSN: 0022-1759, DOI: 10.1016/0022-1759(94)00305-G *
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KOSE ET AL., SCI. IMMUNOL., vol. 4, 2019, pages eaaw6647
KRISKY ET AL., GENE THER, vol. 5, 1998, pages 1517
LEFRANC ET AL., DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55
LEFRANC, M.-P.LEFRANC, G.: "Immunogenetics", vol. 882, 2012, HUMANA PRESS, SPRINGER, article "Human Gm, Km and Am allotypes and their molecular characterization: a remarkable demonstration of polymorphism", pages: 635 - 680
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI ET AL., WILERY INTERDISCIP REV. NANOMEDNANOBIOTECHNOL., vol. 77, no. 2, 2019, pages el530
LI, JQ.ZHANG, ZR.ZHANG, HQ. ET AL.: "Intranasal delivery of replicating mRNA encoding neutralizing antibody against SARS-CoV-2 infection in mice", SIG TRANSDUCT TARGET THER, vol. 6, 2021, pages 369, Retrieved from the Internet <URL:https://doi.org/10.1038/s41392-021-00783-1>
LIU ET AL., MABS, vol. 6, no. 5, 2014, pages 1145 - 1154
MARATEA ET AL., GENE, vol. 40, 1985, pages 39 - 46
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MATES ET AL., NAT. GENET., vol. 41, 2009, pages 153
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
MURPHY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 8258 - 8262
MUTHUMANI ET AL., HUM VACCIN IMMUNOTHER, vol. 9, 2013, pages 2253 - 2262
MUTHUMANI ET AL., J INFECT DIS., vol. 214, no. 3, 2016, pages 369 - 378
NANCEMEIER, ACS CENT. SCI., vol. 7, no. 5, 2021, pages 748 - 756
NORTH ET AL., J MOL BIOL., vol. 406, 2011, pages 228 - 56
PALMBERGER ET AL., J. BIOTECHNOL., vol. 153, no. 3-4, 2011, pages 160 - 166
PARDI ET AL., J CONTROLRELEASE, 2015, pages 217345 - 351
PLUCKTHUN, A, BIO/TECHNOLOGY, vol. 9, 1991, pages 545 - 551
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
RAVETCH, J. VKINET, J. P., ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
SABNIS ET AL., MOL. THER., vol. 26, 2018, pages 1509 - 1519
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
SCATCHARD, ANN. N.Y. ACAD. SCI., vol. 51, 1949, pages 660
SHIELDS, R. L. ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 6591 - 6604
SPIESS ET AL., MOL. IMMUNOL., vol. 67, no. 2, 2015, pages 95
SU CHIA-CHENG ET AL: "Effects of antibodies against human parvovirus B19 on angiogenic signaling", MOLECULAR MEDICINE REPORTS, 9 January 2020 (2020-01-09), GR, XP093077919, ISSN: 1791-2997, DOI: 10.3892/mmr.2020.10921 *
SUZUKI ET AL., CLIN. CANCER RES., vol. 13, no. 6, 2007, pages 1875 - 82
THESS ET AL., MOL THER, vol. 23, 2015, pages 1456 - 1464
THRAN ET AL., EMBO MOL MED, vol. 9, no. 10, 2017, pages 1434 - 1448
TRAGGIAI ET AL., NAT MED, 2004
URLAUB ET AL., PNAS, vol. 77, 1980, pages 4216
VAN DE WINKEL, J. G., ANDERSON, C. L., J. LEUKOC. BIOL., vol. 49, 1991, pages 511 - 524
VAN HOECKEROOSE, J. TRANSLATIONALMED, vol. 17, 2019, pages 54, Retrieved from the Internet <URL:https://doi.org/10.1186/s12967-019-1804-8>
VERHOEYEN ET AL., METHODS MOL. BIOL., vol. 506, 2009, pages 97
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WALCHLI ET AL., PLOS ONE, vol. 6, 2011, pages 327930
WARD, E. S.GHETIE, V, THER. IMMUNOL., vol. 2, 1995, pages 77 - 94
WILSON, SCIENCE, vol. 295, 2002, pages 2103
WINES, B.D. ET AL., J. IMMUNOL., vol. 164, 2000, pages 5313 - 5318
WOLFF ET AL., CANCER RES, vol. 55, 1993, pages 2560
YAZAKIWU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268
ZHANG ET AL., FRONT. IMMUNOL., 2019
ZHAO ET AL., J. IMMUNOL., vol. 174, 2005, pages 4415

Similar Documents

Publication Publication Date Title
EP3872091B1 (fr) Anticorps contre le sars-cov-2
WO2021203053A1 (fr) Immunothérapie ciblant une région conservée dans des coronavirus sras
AU2021268361A1 (en) Antibodies against SARS-CoV-2
WO2021211775A1 (fr) Anticorps dirigés contre le sras-cov-2 et leurs procédés d&#39;utilisation
US20240092876A1 (en) Broadly neutralizing antibodies against influenza neuraminidase
WO2022204202A1 (fr) Anticorps qui se lient à de multiples sarbecovirus
EP4247844A1 (fr) Anticorps contre les virus de la grippe a
US20240141021A1 (en) Anti-influenza antibodies and combinations thereof
WO2023245078A1 (fr) Anticorps anti-parvovirus et leurs utilisations
WO2024118998A2 (fr) Anticorps anti-sars-cov-2 modifiés et leurs méthodes d&#39;utilisation
WO2024006472A1 (fr) Anticorps qui se lient à de multiples sarbecovirus
WO2024112818A1 (fr) Anticorps anti-sars-cov-2 modifiés et leurs utilisations
WO2023230445A2 (fr) Anticorps largement neutralisants dirigés contre la neuraminidase de la grippe
TW202417477A (zh) 與多種嚴重急性呼吸道症候群β型冠狀病毒結合之抗體
WO2024026411A1 (fr) Anticorps neutralisants à large spectre contre les paramyxovirus rsv et mpv

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23741551

Country of ref document: EP

Kind code of ref document: A1