WO2023235764A1 - Anti-cd20 antibody compositions - Google Patents

Anti-cd20 antibody compositions Download PDF

Info

Publication number
WO2023235764A1
WO2023235764A1 PCT/US2023/067711 US2023067711W WO2023235764A1 WO 2023235764 A1 WO2023235764 A1 WO 2023235764A1 US 2023067711 W US2023067711 W US 2023067711W WO 2023235764 A1 WO2023235764 A1 WO 2023235764A1
Authority
WO
WIPO (PCT)
Prior art keywords
population
antibody
infusion
glycans
composition
Prior art date
Application number
PCT/US2023/067711
Other languages
French (fr)
Inventor
George Costas Avgerinos
Patrick Michael Hossler
Jill A. Myers
Yune Z. Kunes
Sri Hari Pandurang Miskin
Michael Weiss
Peter Sportelli
Original Assignee
Tg Therapeutics, Inc.
Laboratoire Francais Du Fractionnement Et Des Biotechnologies
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 Tg Therapeutics, Inc., Laboratoire Francais Du Fractionnement Et Des Biotechnologies filed Critical Tg Therapeutics, Inc.
Publication of WO2023235764A1 publication Critical patent/WO2023235764A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present disclosure is in the field of recombinant anti-CD20 antibodies, methods of producing such antibodies, and uses of such antibodies.
  • Therapeutic monoclonal antibodies (mAbs) produced in mammalian cells are heterogeneous as a result of post-translational modifications (PTMs). PTMs can occur during mAb production, purification, storage, and post-administration. PTMs are therapeutic mAb product quality attributes (PQAs). Controlling PQAs within predefined acceptance criteria is vital to the biopharmaceutical industry because it ensures consistent product quality and reduces potential impacts on drug safety and efficacy (Xu, X. el al., Journal of Applied Bioanalysis 3(2):21-5 (2017)).
  • PTMs vary from chain additions, such as N- and O- linked glycosylation, glycation, cysteinylation and sulfation; chain trimming, such as C-terminal lysine clipping; amino acid modifications such as cyclization (into a N-terminal pyroglutamic acid), deamidation, oxidation, isomerization and carbamylation; to disulfide scrambling of hinge region interchain disulfide bonds.
  • Each antibody can therefore give rise to a myriad of distinct antibody molecules with large activity and potency differences.
  • post-translational modifications of antibodies have been observed and studied for decades, the full impact of the microheterogeneity is yet to be further studied.
  • PTMs can impact antibody functions, for example, pharmacokinetics and pharmacodynamics properties and clinical efficacy.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising about 20 to 40% fucosylated glycans and optionally about 10 to 20% galactosylated glycans.
  • the N-glycan profile comprises 23% to 36% fucosylated glycans, optionally about 30% fucosylated glycans. In some embodiments, the N-glycan profile comprises 16% to 18% galactosylated glycans, optionally about 17% galactosylated glycans.
  • the relative abundance of fucosylated glycans is the percent of fucosylated glycans among all glycans in the N-glycan profile.
  • the relative abundance of galactosylated glycans is the percent of galactosylated glycans among all glycans in the N-glycan profile.
  • the N-glycan profile comprises 12% to 30% bisecting N- glycans, optionally about 18% bisecting N-glycans.
  • the bisecting N- glycans comprise one or more of GOB, G0FB, G1FB, G2FBS1, and G2FBS2.
  • the population of anti-CD20 antibody proteins has an N-glycan profile comprising less than 5% sialylated glycans. In some embodiments, the N-glycan profile comprises less than 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycan. In some embodiments, the N-glycan profile comprises no detectable amount of sialylated glycan. [0011] In some embodiments, the population of anti-CD20 antibody proteins has an N- glycan profile comprising 0.1% to 1.5% Man5 N-glycan. In some embodiments, the N-glycan profile comprises 0.4% to 0.7% Man5 N-glycan. In some embodiments, the N-glycan profile comprises about 0.6% Man5 N-glycan. In some embodiments, Man5 N-glycan is the only high mannose species in the N-glycan profile.
  • the population of anti-CD20 antibody proteins comprises 0.20 to 0.40 mol isoaspartate per mol protein. In some embodiments, the population of anti-CD20 antibody proteins comprises 0.25 to 0.35 mol isoaspartate per mol protein.
  • glutamate at position 1 of the heavy chain is a pyroglutamate and glutamate at position 1 of the light chain is a pyroglutamate.
  • the population of anti-CD20 antibody proteins has an N- glycan profile comprising a relative abundance ratio of 0.1 to 0.15 G1 to GO N-glycans. In some embodiments, the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.5 to 0.9 GIF to G1 N-glycans.
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance ranges:
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
  • the population of anti-CD20 antibody proteins further comprises at least three, four or five N-glycans within the following relative abundance ranges:
  • the N-glycan profile of the population of anti-CD20 antibody proteins is determined using a method comprising: (a) incubating the population of anti-CD20 antibody proteins with an enzyme, wherein the enzyme catalyzes releasing of the N-glycans from the anti-CD20 antibody; (b) measuring the relevant abundance of the released N-gylcans using one or more methods selected from chromatography, mass spectrometry, capillary electrophoresis, and the combination thereof.
  • the method further comprises after step (a) and before step (b) the following steps: (c) purifying the N-glycans; and (d) labeling the N-glycans with a fluorescent compound.
  • the enzyme is PNGase F.
  • the fluorescent compound is 2-aminobenzamide (2- AB).
  • less than 10% of the anti-CD20 antibody proteins in the population is non-glycosylated.
  • less than 5% of the anti-CD20 antibody proteins in the population is non-glycosylated.
  • less than 1% of the anti- CD20 antibody proteins in the population is non-glycosylated.
  • the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
  • the population of anti-CD20 antibody proteins comprises secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
  • the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
  • the population of anti-CD20 antibody proteins further comprises one or more of the following post-translational modifications at the specified abundance:
  • the one or more of the post-translational modifications are measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS).
  • the population has an amount of total protein of 25.5-25.8 mg/mL as measured by absorbance at 280 nm.
  • the anti-CD20 antibody proteins in the population induces greater cytotoxicity in a cell-based antibody-dependent cellular cytotoxicity (ADCC) assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
  • ADCC antibody-dependent cellular cytotoxicity
  • the population has a relative potency of 90 to 163% in a cellbased ADCC assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of 78% to 116% or 73% to 128% in a cell-based complement dependent cytotoxicity (CDC) assay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of 92 to 118% or 82 to 138% in a cellbased CD20 binding activity bioassay compared to that of a commercial reference standard. In some embodiments, the population has a KD value 30 to 70 nM in an FcyRIIIa-158V binding assay as measured by surface plasmon resonance.
  • CDC complement dependent cytotoxicity
  • the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In some embodiments, the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab. In some embodiments, the population has a relative potency of 88 to 113% or 86 to 116% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard. In some embodiments, the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard.
  • the population has 99.2 to 99.9% monomers as detected by size exclusion chromatography (SEC). In some embodiments, the population has 0.1 to 0.8% dimers as detected by SEC. In some embodiments, the population has undetectable level of aggregates as detected by SEC; and/or undetectable level of fragments as detected by SEC. [0030] In some embodiments, the population has 93.6 to 95.9% IgG after purification by non-reduced capillary gel electrophoresis (CGE). In some embodiments, the population has 0.1 to 0.3% high molecular weight species (HMWS) after purification by non-reduced CGE.
  • CGE capillary gel electrophoresis
  • HMWS high molecular weight species
  • the population has 0.7 to 1.2% free light chain (LC) after purification by non-reduced CGE. In some embodiments, the population has 97.7 to 98.0% heavy chain plus light chain species (HC + LC) after purification by reduced CGE.
  • LC free light chain
  • HC + LC heavy chain plus light chain species
  • the population has 20 to 25% acidic isoforms as detected by imaged capillary isoelectric focusing (iCIEF). In some embodiments, the population has 50 to 60% main isoforms as detected by iCIEF. In some embodiments, the population has 20 to 30% basic isoforms as detected by iCIEF. In some embodiments, the population has an average molar ratio of free thiol to anti-CD20 antibody of about 2.0 to 2.2.
  • the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of the N-terminal residue. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 5 N- terminal residues. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 10 N-terminal residues. In some embodiments, terminal lysine amino acid residue of the heavy chain in the anti-CD20 antibody in the population is truncated.
  • compositions described herein comprising the compositions described herein, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 10 mg/mL to 50 mg/mL. In some embodiments, the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL.
  • the pharmaceutical formulation further comprises one or more of the following: sodium chloride, trisodium citrate dehydrate, polysorbate 80, and hydrochloric acid.
  • the pharmaceutical formulation comprises about 9.0 mg/mL of sodium chloride, about 7.4 mg/mL of trisodium citrate dehydrate, about 0.7 mg/mL of polysorbate 80, and/or about 0.4 mg/mL of hydrochloric acid.
  • the anti-CD20 antibody is present in a single dosage form.
  • compositions comprising:
  • compositions described herein wherein the composition comprises a single dosage form of the population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL,
  • populations of anti-CD20 antibody proteins or pharmaceutical formulations described herein wherein the single batch comprises at least 100 g, at least 120 g, or at least 150 g of the anti-CD20 antibody proteins.
  • populations of anti-CD20 antibody proteins or pharmaceutical formulations described herein produced in a 15,000 L or 20,000 L bioreactor.
  • MS multiple sclerosis
  • the composition or the pharmaceutical formulation is administered as i) a first infusion at a dose of about 150 mg of the the anti-CD20 antibody protein, ii) a second infusion two weeks later at a dose of about 450 mg of the the anti-CD20 antibody protein, and iii) subsequent infusions every 24 weeks or six months at a dose of about 450 mg of the the anti-CD20 antibody protein.
  • composition or the pharmaceutical formulation results in one or more of the following pharmacokinetic parameters:
  • a Cavg is between 6,437 ng/mL and 11,443 ng/mL.
  • composition or the pharmaceutical formulation results in one or more of the following pharmacokinetic parameters:
  • the method comprises a treatment period of at least 96 weeks.
  • the subject has been pre-medicated with a corticosteroid 30-60 minutes prior to administration of the composition or the pharmaceutical formulation.
  • the corticosteroid is methylprednisone or dexamethasone.
  • the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
  • the subject has been pre-medicated with an antihistamine 30- 60 minutes prior to administration of the composition or the pharmaceutical formulation.
  • the antihistamine is diphenhydramine HC1.
  • the diphenhydramine HC1 is administered at a dose of about 25-50 mg.
  • the subject has been pre-medicated with an antipyretic 30-60 minutes prior to administration of the composition or the pharmaceutical formulation.
  • the antipyretic is acetaminophen or an antipyretic bioequivalent thereto.
  • the subject has an Expanded Disability Status Scale (EDSS) score of from 0 to 5.5 prior to treatment.
  • EDSS Expanded Disability Status Scale
  • MS multiple sclerosis
  • NED A no evidence of disease activity
  • administration of the composition or the pharmaceutical formulation results in NED A in the subject 24 weeks after the administration.
  • the lymphocyte count is normalized by day 8 of the administration.
  • the MS is a relapsing form of MS (RMS).
  • ARR annualized relapse rate
  • MS multiple sclerosis
  • the effective amount of the composition or the pharmaceutical formulation is sufficient to result in an ARR of 0.091 or an ARR of 0.076.
  • duration of the second infusion, the first subsequent infusion, and the one or more subsequent infusions of the anti-CD20 antibody protein is about one hour.
  • methods of treating relapsing forms of multiple sclerosis (MS) in a subject in need thereof by administering to the subject an effective amount of a composition or pharmaceutical formulation described herein comprising: administering an intravenous infusion of the composition or the pharmaceutical formulation in a multi-infusion dosage regimen, the dosage regimen comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody protein at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody protein at about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about
  • the method further comprises pre-medicating the subject with a corticosteroid and an antihistamine, 30-60 minutes prior to administration of the composition or the pharmaceutical formulation.
  • the corticosteroid is methylprednisone or dexamethasone.
  • the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
  • the intravenous infusion of the composition or the pharmaceutical formulation is prepared in 250 mb of 0.9% Sodium Chloride Injection.
  • the first subsequent infusion is at about 24 weeks from the first infusion. In some embodiments, the one or more subsequent infusions is at about 24 weeks from the prior infusion. In some embodiments, the first subsequent infusion is at about 6 months from the first infusion. In some embodiments, the one or more subsequent infusions is at about 6 months from the prior infusion.
  • duration of the first infusion of the anti-CD20 antibody protein is about four hours.
  • the first infusion of the anti-CD20 antibody protein is infused at a rate of 10 mb per hour for the first 30 minutes; 20 mb per hour for the next 30 minutes; 35 mb per hour for the next hour; and 100 mb per hour for the remaining two hours.
  • the second infusion, the first subsequent infusion, and the one or more subsequent infusion of the anti-CD20 antibody protein is infused at a rate of 100 mb per hour for the first 30 minutes, and 400 mb for the remaining 30 minutes.
  • the multi-infusion dosage regimen of the anti-CD20 antibody protein alleviates or delays progression of the symptoms of MS.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of gadolinium-enhancing T1 lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of new and enlarging T2 hyperintense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased no evidence of disease activity (NED A) status, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • NED A no evidence of disease activity
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased Confirmed Disability Improvement (CDI), as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • CDI Confirmed Disability Improvement
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an increased Multiple Sclerosis Functional Composite (MSFC) score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • MSFC Multiple Sclerosis Functional Composite
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved timed 25-Foot Walk (T25FW) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved 9-Hole Peg test (9-HPT) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • 9-HPT 9-Hole Peg test
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a significant reduction in both volume and number of new T1 hypointense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • Also provided herein are methods of treating multiple sclerosis wherein the methods comprise administering a therapeutically effective amount of a pharmaceutical formulation comprising a population of anti-CD20 antibody proteins to a subject in need thereof, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein administration of the pharmaceutical formulation results in no evidence of disease activity (NED A) in the subject 24 weeks after the administration. In some embodiments, administration of the pharmaceutical formulation results in NED A in the subject 24-96 weeks after the administration.
  • Also disclosed herein are methods of treating multiple sclerosis wherein the methods comprise administering a therapeutically effective amount of a pharmaceutical formulation comprising a population of anti-CD20 antibody proteins to a subject in need thereof, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein administration of the pharmaceutical formulation results in a transient decrease in lymphocyte count in the subject.
  • the lymphocyte count is normalized by day 8 of the administration.
  • the population of anti- CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans.
  • the multiple sclerosis is a relapsing form of MS (RMS).
  • the RMS comprises a clinically isolated syndrome ("CIS”); relapsing-remitting MS ("RRMS”); or active secondary progressive MS (“SPMS”).
  • CIS clinically isolated syndrome
  • RRMS relapsing-remitting MS
  • SPMS active secondary progressive MS
  • the subject is diagnosed with RMS in accordance to McDonald Criteria (2010), or by another suitable method known by one skilled in the art.
  • the subject is a human.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 35% fucosylated glycans. [0080] In some embodiments, the N-glycan profile comprises 28 to 33% fucosylated glycans.
  • the N-glycan profile comprises about 30% fucosylated glycans. [0081] In some embodiments, the N-glycan profile comprises 16 to 18% galactosylated glycans. In some embodiments, the N-glycan profile comprises about 17% galactosylated glycans.
  • the relative abundance of fucosylated glycans is the percent of fucosylated glycans among all glycans in the N-glycan profile.
  • the relative abundance of galactosylated glycans is the percent of galactosylated glycans among all glycans in the N-glycan profile.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising at least about 10% bisecting N-glycans.
  • the N-glycan profile comprises 12% to 30% bisecting N- glycans. In some embodiments, the N-glycan profile comprises about 18% bisecting N-glycans. In some embodiments, the bisecting N-glycan comprises one or more of GOB, G0FB, G1FB, G2FBS1, and G2FBS2.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising less than 5% sialylated glycans.
  • the N-glycan profile comprises less than 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycan. In some embodiments, the N-glycan profile comprises no detectable amount of sialylated glycan.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 5 to 15% GOB N-glycan.
  • the N-glycan profile comprises 9 to 11% GOB N-glycan. In some embodiments, the N-glycan profile comprises about 10% GOB N-glycan.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 0.1% to 1.5% Man5 N-glycan.
  • the N-glycan profile comprises 0.4% to 0.7% Man5 N-glycan. In some embodiments, the N-glycan profile comprises about 0.6% Man5 N-glycan. In some embodiments, Man5 N-glycan is the only high mannose species in the N-glycan profile.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins comprises 0.20 to 0.40 mol isoaspartate per mol protein.
  • the population of anti-CD20 antibody proteins comprises 0.25 to 0.35 mol isoaspartate per mol protein.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the glutamate at position 1 of the heavy chain is a pyroglutamate and wherein the glutamate at position 1 of the light chain is a pyroglutamate.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.1 to 0.15 G1 to GO N-glycans.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.5 to 0.9 GIF to G1 N-glycans.
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance range: (a) 0.3% to 2% G0-GN; (b) 0.1% to 2% G0F-GN; (c) 0.1% to 1% Gl-GN; (d) 5% to 20% GOB; (e) 5% to 30% G0F; (f) 0.1% to 1.5% Man5; (g) 1% to 15% G0FB; (h) 1% to 13% Gl; (i) 0.5% to 10% GF; (j) 0.5% to 6% GIB; (k) 0.5% to 12% GIF; (1) 0.1% to 3% GIF’; (m) 0.1% to 3% G1FB; (n) 0.1% to 2% G2; and (o) 0.1% to 2% G2F.
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance range: (a) 0.8% to 1.1% G0-GN; (b) 0.5% to 1.1% G0F-GN; (c) 0.3% to 0.6% Gl-GN; (d) 9.5% to 14.1% GOB; (e) 12.8% to 19.7% G0F; (f) 0.4% to 0.7% Man5; (g) 5.1% to 7.0% G0FB; (h) 5.7% to 6.4% Gl; (i) 2.7% to 3.3% GF; (j) 1.4% to 2.0% GIB; (k) 2.6% to 4.2% GIF; (1) 1.1% to 1.6% GIF’; (m) 1.1% to 1.8% G1FB; (n) 0.5% to 0.7% G2; and (o) 0.3% to 0.5% G2F.
  • N-glycans within the following relative abundance range: (a) 0.8% to 1.1% G0-GN; (b) 0.5% to 1.1% G0F-GN; (c) 0.3% to 0.6% Gl
  • the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance: (a) 0.9% G0-GN; (b) 0.8% G0F-GN; (c) 0.5% Gl-GN; (d) 10.9% GOB; (e) 17.0% G0F; (f) 0.6% Man5; (g) 6.0% G0FB; (h) 6.1% Gl; (i) 2.9% GF; (j) 1.6% GIB; (k) 3.2% GIF; (1) 1.3% GIF’; (m) 1.3 G1FB; (n) 0.5% G2; and (o) 0.3% G2F.
  • the population of anti-CD20 antibody proteins further comprises at least three, four or five N-glycans within the relative abundance or relative abundance range as described herein.
  • the N-glycan profile of the population of anti-CD20 antibody proteins is determined using a method comprising: (a) incubate the population of anti-CD20 antibody proteins with an enzyme, wherein the enzyme catalyzes releasing of the N-glycans from the anti-CD20 antibody; (b) measure the relevant abundance of the released N-gylcans using one or more methods selected from chromatography, mass spectrometry, capillary electrophoresis, and the combination thereof.
  • the enzyme is PNGase F.
  • the method further comprises after step (a) and before step (b) the following steps: (c) purify the N-glycans; and (d) label the N-glycans with a fluorescent compound.
  • the fluorescent compound is 2-aminobenzamide (2-AB).
  • less than 10% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 5% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 1% of the anti- CD20 antibody proteins in the population is non-glycosylated.
  • the N-glycan profile of the population of anti-CD20 antibody proteins is substantially as shown in Figure 2.
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) 8.0% to 10.0% a-helix; (b) 32.0% to 36.0% Anti-parallel
  • the population of anti-CD20 antibody proteins comprises secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) 8.0% to 10.0% a-helix; (b) 32.0% to 36.0% Anti-parallel
  • the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) about 9.0% a-helix; (b) about 33.0% Anti-parallel
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain (“HC”) comprising the amino acid sequence of SEQ ID NO: 1 and a light chain (“LC”) comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins further comprises one or more of the following post-translational modifications at the specified abundance:
  • HC heavy chain
  • LC light chain
  • the population of anti-CD20 antibody proteins comprises two, three, four, five or more of the post translational modifications.
  • the population of anti-CD20 antibody proteins comprises the following post-translational modification at the specified abundance:
  • the population of anti-CD20 antibody proteins comprises the following post-translational modification at the specified abundance:
  • one or more of the post-translational modifications are measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS).
  • the deamidation is measured by isoaspartate detection or peptide mapping using LC-MS.
  • compositions comprising a population of anti-CD20 antibody proteins described herein, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain (“HC”) comprising the amino acid sequence of SEQ ID NO: 1 and a light chain (“LC”) comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has the following properties:
  • the population has an amount of total protein of 25.5-25.8 mg/mL as measured by absorbance at 280 nm. In some embodiments, the population has an amount of total protein of about 25.6 mg/mL as measured by absorbance at 280 nm. [00114] In some embodiments, the population induces greater cytotoxicity in a cell-based antibody-dependent cellular cytotoxicity (ADCC) assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
  • ADCC antibody-dependent cellular cytotoxicity
  • the population has a relative potency of 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 117% in a cell-based ADCC assay compared to a commercial reference standard.
  • the cell-based ADCC assay uses effector cells selected from CD 16 effector cells and primary NK cells. In some embodiments, the population performs in a cell-based ADCC using CD 16 effector cells at more than 100% of that of a commercial reference standard.
  • the population exhibits greater B cell depletion activity in a human whole blood B cell depletion assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
  • the population has a relative potency of 78% to 116% or 73% to 128% in a cell-based complement dependent cytotoxicity (CDC) assay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of about 91% in a cell-based CDC assay compared to that of a commercial reference standard. [00117] In some embodiments, the population has a relative potency of 92 to 118% or 82 to 138% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of about 109% in a cellbased CD20 binding activity bioassay compared to that of a commercial reference standard.
  • CDC complement dependent cytotoxicity
  • the population has a KD value 30 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In some embodiments, the population has a KD value about 59 nM in an FcyRIIIa- 158 V binding assay as measured by surface plasmon resonance.
  • the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In some embodiments, the population has a KD value 760 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab.
  • the population has a relative potency of 88 to 113% or 86 to 116% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of about 99% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 115% in a CD 16 activity assay compared to a commercial reference standard.
  • the population of anti-CD20 antibody proteins has a purification profile as follows:
  • the population has 99.2 to 99.9% monomers as detected by size exclusion chromatography (SEC). In some embodiments, the population has 0.1 to 0.8% dimers as detected by SEC. In some embodiments, the population has undetectable level of aggregates as detected by SEC. In some embodiments, the population has undetectable level of fragments as detected by SEC.
  • SEC size exclusion chromatography
  • the population has 93.6 to 95.9% IgG after purification by non-reduced capillary gel electrophoresis (CGE). In some embodiments, the population has 0.1 to 0.3% high molecular weight species (HMWS) after purification by non-reduced CGE. In some embodiments, the population has 0.7 to 1.2% free light chain (LC) after purification by non-reduced CGE.
  • CGE capillary gel electrophoresis
  • HMWS high molecular weight species
  • LC free light chain
  • the population has 97.7 to 98.0% heavy chain plus light chain species (HC + LC) after purification by reduced CGE.
  • the population of anti-CD20 antibody proteins has a distribution of charged isoforms as follows:
  • the population has 20 to 25% acidic isoforms as detected by imaged capillary isoelectric focusing (iCIEF). In some embodiments, the population has 50 to 60% main isoforms as detected by iCIEF. In some embodiments, the population has 20 to 30% basic isoforms as detected by iCIEF.
  • iCIEF imaged capillary isoelectric focusing
  • the population has an average molar ratio of free thiol to anti- CD20 antibody of about 2.0 to 2.2.
  • the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of the N-terminal residue. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 5 N- terminal residues. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 10 N-terminal residues.
  • the terminal lysine amino acid residue of the heavy chain in the anti-CD20 antibody in said population is truncated.
  • the administration of the anti-CD20 antibody to a human patient results in one or more of the following pharmacokinetic parameters: (a) an AUC between 2,160 pg/mL and 3,840 pg/mL; (b) a Cmax between 118,011 ng/mL and 159,989 ng/mL; (c) a Cmin between 40 ng/mL and 375 ng/mL; and (d) a Cavg is between 6,437 ng/mL and 11,443 ng/mL, and wherein the anti-CD20 antibody is administered as i) a first infusion at a dose of about 150 mg, ii) a second infusion two weeks later at a dose of about 450 mg, and iii) subsequent infusions every six months at a dose of about 450 mg.
  • the administration of the anti-CD20 antibody to a human patient results in one or more of the following pharmacokinetic parameters: (a) an AUC about 3,000 pg/mL; (b) a Cmax about 139,000 ng/mL; (c) a Cmin about 139 ng/mL; and (d) a Cavg about 8,940 ng/mL.
  • the antibody proteins of the population are present in a single dosage form.
  • compositions described herein comprising compositions described herein, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL.
  • compositions comprising anti-CD20 antibody described herein, wherein the anti-CD20 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the pharmaceutical formulation comprises one or more of the following: sodium chloride, trisodium citrate dehydrate, polysorbate 80, and hydrochloric acid.
  • the pharmaceutical formulation comprises about 9.0 mg/mL of sodium chloride. In some embodiments, the pharmaceutical formulation comprises about 7.4 mg/mL of trisodium citrate dehydrate. In some embodiments, the pharmaceutical formulation comprises about 0.7 mg/mL of polysorbate 80. In some embodiments, the pharmaceutical formulation comprises about 0.4 mg/mL of hydrochloric acid.
  • Also provided here are methods of treating an autoimmune disease wherein the method comprises administering the compositions described herein to a subject in need thereof, and wherein the autoimmune disease is selected from the group consisting of psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, osteoporosis, eczema, allogenic or xenogenic transplantation, graft rejection, graft- versus-host disease, lupus erythematosus, inflammatory disease, type 1 diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis, myasthenia gravis, autoimmune hemolytic anemia, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic con
  • the multiple sclerosis is a relapsing form of MS.
  • the relapsing form of MS is selected from clinically isolated syndrome (CIS), relapsing-remitting MS (RRMS) and active secondary progressive MS (SPMS).
  • the relapsing form of MS is a clinically isolated syndrome (CIS).
  • the relapsing form of MS is relapse-remitting multiple sclerosis (RRMS).
  • the relapsing form of MS is active secondary progressive multiple sclerosis (SPMS).
  • compositions described herein are administered to a subject in need thereof.
  • the neoplastic disease is acute B lymphoblastic leukaemia, B- cell lymphoma, mature B-cell lymphoma, including B-type Chronic Lymphocytic Leukaemia (B-CLL), small B-cell lymphoma, B-cell prolymphocytic leukaemia, lymphoplasmocytic lymphoma, mantle cell lymphoma, follicular lymphoma, marginal zone MALT-type lymphoma, lymph node marginal zone lymphoma with or without monocytoid B cells, splenic marginal zone lymphoma (with or without villous lymphocytes), tricholeucocytic leukaemia, diffuse large B- cell lymphoma, or Burkitt's lymphoma.
  • B-CLL B-type Chronic Lymphocytic Leukaemia
  • small B-cell lymphoma B-cell prolymphocytic leukaemia
  • lymphoplasmocytic lymphoma lymphoplasmoc
  • TG-1101 TG Therapeutics, Inc.
  • N-glycan fraction (ii) analyzing the N-glycan fraction to determine if one or more N-glycans are the following N- glycans within the following relative abundance range: (a) 0.3% to 2% G0-GN; (b) 0.1% to 2% G0F-GN; (c) 0.1% to 1% Gl-GN; (d) 5% to 20% GOB; (e) 5% to 30% G0F; (f) 0.1% to 1.5% Man5; (g) 1% to 15% G0FB; (h) 1% to 13% Gl; (i) 0.5% to 10% Gl’; (j) 0.5% to 6% GIB; (k) 0.5% to 12% GIF; (1) 0.1% to 3% GIF’; (m) 0.1% to 3% G1FB; (n) 0.1% to 2% G2; and (o) 0.1% to 2% G2F.
  • the method comprises analyzing the N-glycan fraction to determine if one or more N-glycans are the following N-glycans within the following relative abundance range:
  • the method comprises analyzing the N-glycan fraction to determine if one or more N-glycans are in the following relative abundance: (a) 0.9% G0-GN;
  • compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance: (a) 0.9% GO-GN; (b) 0.8% GOF-GN; (c) 0.5% Gl-GN; (d) 10.9% GOB; (e) 17.0% G0F; (f) 0.6% Man5; (g) 6.0% G0FB; (h) 6.1% Gl; (i) 2.9% G1 ’ ; (j) 1-6% GIB; (k) 3.2% GIF; (1) 1.3% GIF’; (m) 1.3 G1FB; (n) 0.5% G2; and (o) 0.3% G2F, and wherein the population of anti-
  • the rat hybridoma cell is YB2/0 cell.
  • Figure 1 illustrates the structure and abbreviations for various N-glycans.
  • Figure 2 illustrates the glycosylation profile of a sample of anti-CD20 antibody proteins provided herein.
  • Figure 3 illustrates the intact MS Spectra of a sample of anti-CD20 antibody proteins provided herein.
  • Figure 4 illustrates antibody-dependent cellular cytotoxicity (ADCC) activity dose response curves using Raji cells and KILR CD16a cells.
  • Figure 5 illustrates ADCC activity dose response curves using Raji cells and primary NK cells.
  • Figure 6 illustrates antibody-dependent cellular phagocytosis (ADCP) activity dose response curves.
  • Figure 7 illustrates complement dependent cytotoxicity (CDC) activity dose response curves.
  • Figure 8 illustrates CD20 binding dose response curves.
  • Figure 9 illustrates CD20 binding dose response curves using FACS.
  • Figure 10 illustrates Clq binding dose response curves.
  • Figures 11A-C illustrate human whole blood B cell depletion from three donors. B cell depletion calculated based on data using CD 19 as B cell marker.
  • Figure 12 illustrates the Goodness-of-Fit (GOF) diagnostics of the TG-1101 final model.
  • Figure 13 illustrates the pcVPC for the TG-1101 final PK model by study.
  • the blue dots are prediction-corrected observed concentrations; the blue lines are the 50th (solid), 5th (dashed), and 95th (dashed) percentiles of observed concentrations; and the black lines are the 50th (solid), 5th (dashed), and 95th (dashed) percentiles of simulations.
  • the gray bands are the 95% Pls for the corresponding black lines based on 500 simulations.
  • the short yellow lines indicate bin intervals.
  • the numbers 201,301,302 and 101-304-703 represent the study numbers, TG1101-RMS201, TG1101-RMS301 and TG1101-RMS302 and the studies in the previous analysis set in subjects with hematologic malignancies, respectively.
  • Figure 14 illustrates the forest plot of covariate effects on TG-1101 drug exposure.
  • the first and second dashed vertical lines correspond to ratios of 0.8 and 1.25, respectively.
  • the solid vertical line corresponds to a ratio of 1 and represents a typical subject. Points and whiskers represent the estimate and 90% confidence interval, respectively.
  • the blue-gray horizontal bar shows the range of exposures due to between-subject variability.
  • a typical subject is defined as a male subject from North America/Western Europe with a body weight of 73 kg, is ADA negative and has been on treatment for ⁇ 416 days.
  • ADA antidrug antibody
  • AUCss area under the serum TG-1101 concentration-time curve at steady state
  • BSV between-subject or inter-individual variability
  • CI confidence interval
  • CMAXSS maximum TG-1101 concentration at steady state
  • CMINSS minimum TG-1101 concentration at steady state
  • N/A not applicable.
  • Figure 15 depicts the map of expression vector, HK463-25, containing the immunoglobulin heavy and light chain cDNA sequences of source anti-CD20 antibody, TG- 1101, described herein.
  • Figures 16A-16C depict the onset and maintenance of no evidence of disease activity (NED A) with ublituximab.
  • Figure 16A is a bar graph showing NEDA-3 rates by treatment EPOCH (Weeks 0-24 vs Weeks 24-96). denominator based on participants in the Weeks 24-96 analysis. Pooled post hoc analysis. Modified intention-to-treat population.
  • Figure 16B is a bar graph showing NEDA-3 rates by treatment EPOCH (Weeks 24-48 vs Weeks 48-96). denominator based on participants in the Weeks 48-96 analysis. Pooled post hoc analysis. Modified intention-to-treat population.
  • Figure 16C is a bar graph showing components driving EDA in ublituximab-treated participants 3 .
  • 3 Participants may have >1 component of EDA. Pooled post hoc analysis. Modified intention-to-treat population.
  • CDP confirmed disability progression
  • EDA evidence of disease activity
  • Gd+ gadolinium-enhancing
  • n/e new/enlarging
  • NED A no evidence of disease activity
  • NEDA-3 3-parameter NED A
  • EPOCH combination of etoposide phosphate, prednisone, vincristine sulfate (Oncovin), cyclophosphamide, and doxorubicin hydrochloride (hydroxy daunorubicin).
  • Figures 17A-17C depict characterization of cytopenias with ublituximab in the ULTMATE I and II Phase 3 studies in participants with relapsing multiple sclerosis.
  • Figure 17A is a bar graph showing hematologic laboratory abnormalities in ublituximab-treated participants (all study visits). a (493/543). Percentages are based on number of participants with non-missing baseline and at least 1 postbaseline assessment. Pooled post hoc analysis. Safety population.
  • Figure 17B is a bar graph showing hematologic laboratory abnormalities in ublituximab-treated participants (excluding Day 2). a (l 75/543).
  • Percentages are based on number of participants with non-missing baseline and at least 1 postbaseline assessment. Pooled post hoc analysis. Safety population. Figure 17C is a bar graph showing proportion of ublituximab-treated participants with low lymphocyte count by visit. a (476/523). Pooled post hoc analysis. Safety population. Percentages are based on number of participants in the population/treatment group. BL, baseline.
  • Figures 18A-18B are bar graphs showing the relative reduction in Annualized Relapse Result (ARR) in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples.
  • the ARR results from ULTIMATE I ( Figure 18A) and ULTIMATE II ( Figure 18B) are shown.
  • Figures 19A-19B are bar graphs showing the relative reduction in Total Number of Gadolinium (Gd)+ T1 Lesions on MRI in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 19A) and ULTIMATE II ( Figure 19B) are shown.
  • Figures 20A-20B are bar graphs showing the relative reduction in the number of New or Enlarging T2 Lesions on MRI in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 20A) and ULTIMATE II ( Figure 20B) are shown.
  • Figures 21A-21B are bar graphs showing the improvement in the proportion (%) of patients with No Evidence of Disease Activity (NED A) status in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples.
  • the NEDA results from ULTIMATE I ( Figure 21A) and ULTIMATE II ( Figure 21B) are shown.
  • Figures 22A-22B are graphs comparing the percentage of RMS patients having Confirmed Disability Progression (CDP) in a pre-specified pooled analysis of patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples.
  • Figure 22A shows 12- week CDP and
  • Figure 22B shows 24-week CDP.
  • Figures 23A-23B are graphs comparing the percentage of RMS patients achieving Confirmed Disability Improvement (CDI) in a pre-specified pooled analysis of patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples.
  • Figure 23A shows 12- week CDI
  • Figure 23B shows 24-week CDI.
  • Figures 24A-24B are bar graphs showing significant improvements from baseline to 96 weeks in mean Multiple Sclerosis Functional Composite (MSFC) scores in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 24A) and ULTIMATE II ( Figure 24B) are shown.
  • MSFC Multiple Sclerosis Functional Composite
  • Figures 25A-25B are bar graphs showing significant improvements in mean Timed 25-Foot Walk (T25FW) scores from baseline to 96 weeks in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 25A) and ULTIMATE II ( Figure 25B) are shown.
  • T25FW Timed 25-Foot Walk
  • Figures 26A-26B are bar graphs showing the changes in mean Paced Auditory Serial Additional Test (PAS AT) scores from baseline to 96 weeks in RMS patients administered ublituximab and teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 26A) and ULTIMATE II ( Figure 26B) are shown. The change in mean PASAT scores was similar between the treatment groups.
  • PAS AT Paced Auditory Serial Additional Test
  • Figures 27A-27B are bar graphs showing significant increases in mean 9-Hole Peg Test (9-HPT) scores from baseline to 96 weeks in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 27 A) and ULTIMATE II ( Figure 27B) are shown.
  • 9-HPT 9-Hole Peg Test
  • Figures 28A-28B are graphs showing Percent Brain Volume Change (PBVC) on MRI from week 24 to week 96 (post-hoc analysis) of RMS patients administered ublituximab and teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I ( Figure 28A) and ULTIMATE II ( Figure 28B) are shown.
  • PBVC Percent Brain Volume Change
  • the primary amino acid sequences of such antibodies are provided in Section 7.3.
  • Types of such post-translational modifications and their respective abundance in the anti-CD20 antibody populations are described in Section 7.4.
  • Compositions, including single-batch compositions, of such populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications and ranges of purity are described in Section 7.5(e).
  • Assays for quantifying such post-translational modifications in a population of anti- CD20 antibody proteins are described in Section 7.5.
  • Assays for analyzing populations of anti- CD20 antibody proteins are described in Section 7.5.
  • TG-1101 (TG Therapeutics, Inc.) (also known as ublituximab, UBX, UTX, TG-1101, TGTX-1101, UtuxinTM, LFB-R603, TG20, EMAB603) is the source antibody for the anti-CD20 antibody described herein having a unique glycosylation profile that is produced by the disclosed methods.
  • the source antibody, TG-1101 is a monoclonal antibody that targets epitopes on CD20, e g., IRAHT (SEQ ID NO: 37), and EPAN (SEQ ID NO: 38). See, Fox, E. et al., Mult.
  • the term “population of anti-CD20 antibody proteins” refers to a composition of anti-CD20 antibody proteins that is being tested for the abundance of post- translational modifications.
  • the individual anti-CD20 antibody proteins in a population can comprise similar or different post-translational modifications.
  • a population of anti-CD20 antibody proteins refers to all anti-CD20 antibody proteins that are present in a single dosage form.
  • a population of anti-CD20 antibody proteins refers to all anti-CD20 antibody proteins that are present in a single batch.
  • a population of anti-CD20 antibody proteins is an amount sufficient to determine whether the batch of anti-CD20 antibody proteins, when compared to a reference standard, meets or fails a predetermined acceptable range of comparison value or values.
  • the term “single batch” in the context of anti-CD20 antibody proteins refers to a composition derived from a single production or run from a single bioreactor of a specified volume.
  • the anti-CD20 antibody proteins obtained from a single run of a 15,000L bioreactor can be referred to as a single batch.
  • the bioreactor has a capacity of at least 100; 200; 300; 400; 500; 750; 1,000; 2,000; 3,000; 4,000; 5,000; 7,500; 10,000; 15,000; 20,000; or at least 25,000 L.
  • the term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1 or 2 standard deviations. In certain embodiments, the term “about” or “approximately” means within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.
  • the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2. Sequences are provided below in the Sequence Table.
  • the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 or an amino acid that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99% identical to SEQ ID NO: 1; and a light chain comprising the amino acid sequence of SEQ ID NO:2 or an amino acid that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99% identical to SEQ ID NO:2.
  • the anti-CD20 antibody proteins provided herein comprises the VH CDR1, CDR2, and CDR3 region of sequences SEQ ID NOs: 3, 4, and 5, and the VL CDR1, CDR2, and CDR3 region of sequences SEQ ID NOs: 8, 9, and 10.
  • the anti-CD20 antibody proteins provided herein comprises the VH of SEQ ID NO: 6 and the VL of SEQ ID NO: 11.
  • nucleic acid sequence encoding the heavy chain of the anti-CD20 antibody proteins provided herein comprises the nucleic acid sequence of SEQ ID NO:35. In some aspect, the nucleic acid sequence encoding the light chain of the anti-CD20 antibody proteins provided herein comprises the nucleic acid sequence of SEQ ID NO: 36.
  • the anti-CD20 antibody proteins provided herein bind to the same epitope as TG-1101 (TG Therapeutics, Inc.).
  • the anti-CD20 antibody proteins provided herein are chimeric immunoglobulin G1 (IgGl) anti-CD20 monoclonal antibody proteins, each comprised of a tetrameric assembly from two light chains (213 amino acids) and two heavy chains (448 amino acids).
  • IgGl immunoglobulin G1
  • the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2 and comprise a pyroglutamate at position 1 of light chain and/or heavy chain instead of glutamine, thus resulting in an amino acid sequence of SEQ ID NO: 13 for the heavy chain and/or an amino acid sequence of SEQ ID NO: 14 for the light chain.
  • the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2 and comprise a deletion of the C-terminal lysine at the heavy chain thus resulting in the amino acid sequence of SEQ ID NO: 15.
  • composition or population of anti-CD20 antibody proteins wherein at least 50%, 60%, 70%, 80%, 90%, 95%, or 98% comprises (i) a pyroglutamate at position 1 (instead of glutamine) of the heavy chain, (ii) a pyroglutamate at position 1 (instead of glutamine) of the light chain, and/or (iii) a deletion of the C-terminal lysine of the heavy chain.
  • an anti-CD20 antibody is expressed from one or more nucleic acid sequences encoding a light chain comprising the amino acid sequence of SEQ ID NO: 16.
  • the anti-CD20 antibody compositions provided herein can be described by various post-translational modifications and/or by their three-dimensional conformation (see Section 7.5(e)). The respective levels of the various post-translational modifications can be quantified as described in Section 7.5. Without being bound by theory, these structural properties of the anti- CD20 antibody compositions provided herein result in the biological and clinical properties described below in Sections 7.7 and 7.9.
  • the anti-CD20 antibody compositions produced in vitro have various post- translational modifications. It is understood that each individual anti-CD20 antibody protein may have its own specific pattern of post-translational modifications. To describe the properties of a population of multiple anti-CD20 antibody proteins, the overall presence of a specific post- translational modification can be quantified. Without being bound by theory, the levels of a specific post-translational modification in a population of anti-CD20 proteins can determine the biological and clinical properties of the composition (such as a dose of a pharmaceutical formulation). Without being bound by theory, the post-translational modifications are rendered by the expression in a rat hybridoma cell (e.g., YB2/0 cell) in cell culture.
  • a rat hybridoma cell e.g., YB2/0 cell
  • a type of post-translational modification that can be used to describe the anti-CD20 antibody compositions provided herein is glycosylation.
  • glycosylation Various glycosylations are known.
  • the glycosylation is N-glycosylation.
  • N-glycans that can be present can be any one of the N-glycans shown in Figure 1. Levels of N-glycosylation are discussed in the Section 7.4(a) below and can be quantified using the assays in Section 7.5.
  • a type of post-translational modification that can be used to describe the anti-CD20 antibody compositions provided herein is deamidation.
  • Deamidation is a type of post-translational modification that can be used to describe the anti-CD20 antibody compositions provided herein.
  • N-glycosylation can be present in the anti-CD20 antibody compositions provided herein.
  • the relative distribution of the various N-glycans, or individual sugar residues present in those N-glycans, among the individual anti-CD20 antibody proteins in a population of anti-CD20 antibody proteins can determine the biological and clinical properties (such as the biological and clinical properties discussed in Sections 7.7 and 7.9) of the anti-CD20 antibody composition provided herein.
  • the anti-CD20 antibody composition provided herein can be described by any one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or all of the N-glycans or individual sugar residues described in the following subsections.
  • an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
  • an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
  • an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.3% to about 2% G0-GN, from about 0.8% to about 1.1% G0-GN, or about 0.9% G0-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 2% G0F-GN, from about 0.5% to about 1.1% G0F-GN, or about 0.8% G0F-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 30% to 60% GO, from about 42.5% to 48.8% GO, or about 46.1% GO.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 1% Gl-GN, from about 0.3% to about 0.6% Gl-GN, or about 0.5% Gl-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 5% to about 20% GOB, from about 5% to about 15% GOB, from about 9.5% to about 14.1% GOB, about 10.9% GOB, or about 10% GOB.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 5% to about 30% GOF, from about 12.8% to about 19.7% GOF, or about 17.0% GOF. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 1.5% Man5, from about 0.4% to about 0.7% Man5, or about 0.6% Man5. In some embodiments, Man5 is the only high mannose N-glycan in the N-glycan profile.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 1% to about 15% GOFB, from about 5.1% to about 7.0% GOFB, or about 6.0% GOFB. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 1% to about 13% Gl, from about 5.7% to about 6.4% Gl, or about 6.1% Gl. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 10% Gl ’, from about 2.7% to about 3.3% Gl’, or about 2.9% Gl ’.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 6% GIB, from about 1.4% to about 2.0% GIB, or about 1.6% GIB. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 12% GIF, from about 2.6% to about 4.2% GIF, or about 3.2% GIF. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 3% GIF’, from about 1.1% to about 1.6% GIF’, or about 1.3% GIF’.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 3% G1FB, from about 1.1% to about 1.8% G1FB, or about 1.3 G1FB. In certain embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 2% G2, from about 0.5% to about 0.7% G2, or about 0.5% G2. In some embodiments, the population of anti-CD20 antibody proteins comprises a relative abundance of from an N-glycan profile comprising about 0.1% to about 2% G2F, from about 0.3% to about 0.5% G2F, or about 0.3% G2F.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.3% to about 2% G0-GN, from about 0.1% to about 2% G0F-GN, from about 30% to 60% GO, from about 0.1% to about 1% Gl-GN, from about 5% to about 20% GOB, from about 5% to about 30% G0F, from about 0.1% to about 1.5% Man5, from about 1% to about 15% GOFB, from about 1% to about 13% Gl, from about 0.5% to about 10% Gl’, from about 0.5% to about 6% GIB, from about 0.5% to about 12% GIF, from about 0.1% to about 3% GIF’, from about 0.1% to about 3% G1FB, from about 0.1% to about 2% G2, and from about 0.1% to about 2% G2F.
  • N-glycan profile comprising a relative abundance of from about 0.3% to about 2% G0-GN, from about 0.1% to about 2% G0F-GN, from about 30% to 60% GO,
  • Man5 is the only high mannose N-glycan in the N-glycan profile.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.8% to about 1.1% GO-GN, from about 0.5% to about 1.1% G0F-GN, from about 42.5% to 48.8% GO, from about 0.3% to about 0.6% Gl-GN, from about 9.5% to about 14.1% GOB, from about 12.8% to about 19.7% G0F, from about 0.4% to about 0.7% Man5, from about 5.1% to about 7.0% G0FB, from about 5.7% to about 6.4% Gl, from about 2.7% to about 3.3% Gl’, from about 1.4% to about 2.0% GIB, from about 2.6% to about 4.2% GIF, from about 1.1% to about 1.6% GIF’, from about 1.1% to about 1.8% G1FB, from about 0.5% to about 0.7% G2, and from about 0.3% to about 0.5% G2F.
  • GIF is the only high mannose N-glycan in
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of about 0.9% GO-GN, about 0.8% G0F-GN, about 46.1% GO, about 0.5% Gl-GN, about 10.9% GOB, about 17.0% G0F, about 0.6% Man5, about 6.0% G0FB, about 6.1% Gl, about 2.9% Gl ’, about 1.6% GIB, about 3.2% GIF, about 1.3% GIF’, about 1.3 G1FB, about 0.5% G2, and about 0.3% G2F.
  • Man5 is the only high mannose N-glycan in the N-glycan profile.
  • the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance ratio of about 0.1 to about 0.15 Gl to GO N- glycans. In some embodiments, the population of anti-CD20 antibody proteins comprises an N- glycan profile comprising a relative abundance ratio of about 0.5 to about 0.9 GIF to Gl N- glycans.
  • an anti-CD20 antibody composition has an N-glycan profile as shown in Figure 2.
  • compositions or populations of anti-CD20 antibody proteins provided herein can be described by any one of the prevalence of any of these N-glycans or by any group or by all of these N-glycans.
  • an anti-CD20 antibody composition provided herein comprises between 10-20% galactosylated glycans.
  • Galactosylated glycans are those N-glycans shown in Figure 1 that carry a galactose residue (shown as open circle in Figure 1).
  • Assays for determining the percentage of galactosylation (or galactosylated N-glycans) are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N- glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of galactosylated N-glycans is the percent of galactosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises between at least 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, or at least 18% and at most 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or at most 20% galactosylated glycans.
  • an anti-CD20 antibody composition provided herein comprises between 11% and 19%, 12% and 18%, 13% and 17%, or 14% and 16% of galactosylated glycans.
  • an anti-CD20 antibody composition provided herein can comprise about 17% galactosylated glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • an anti-CD20 antibody composition comprises between 20% to 40% fucosylated glycans; between 23% to 36% fucosylated glycans; between 20% and 35% fucosylated glycans; between 28% and 33% fucosylated glycans; or about 33% fucosylated glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • Fucosylated glycans are those N-glycans shown in Figure 1 that carry a fucose residue (shown as open triangle in Figure 1). Assays for determining the percentage of fucosylation (or fucosylated N-glycans) are described in Section 7.5.
  • a sample or a population of anti- CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of fucosylated N-glycans is the percent of fucosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition comprises between at least 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, or at least 33% and at most 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 39%, 40%, 41%, or at most 42% fucosylated glycans.
  • an anti-CD20 antibody composition provided is characterized by a specified ratio of total fucosylated glycans to total galactosylated glycans (or “Fucose/Galactose ratio”).
  • This Fucose/Galactose ratio can be between 1.5 and 2.1 ; between 1.5 and 2; between 1.5 and 1.9; between 1.5 and 1.8; between 1.6 and 2.1; between 1.7 and 2.1; between 1.8 and 2.8; between 1.6 and 2.0; between 1.7 and 1.9; between 1.6 and 1.8; or the Fucose/Galactose ratio can be about 1.75 (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • Fucosy lated glycans are those N-glycans shown in Figure 1 that carry a fucose residue (shown as open triangle in Figure 1).
  • Galactosylated glycans are those N-glycans shown in Figure 1 that carry a galactose residue (shown as open circle in Figure 1).
  • Assays for determining the percentage of fucosylation (or fucosylated N-glycans) are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of fucosylated N-glycans is the percent of fucosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises at least 10%, 15%, 20%, 25%, or at least 30% bisecting N-glycans; between 10% and 30%, between 12% and 30%, between 12% and 25%, between 12% and 20%, between 15% and 30%, between 15% and 25%, between 15% and 20%, between 18% and 30%, or between 18% and 25% bisecting N-glycans.
  • Bisecting N-glycans are those N-glycans shown in Figure 1 that have a third GlcNAc attached to the mannose residue closest to the protein backbone (shown as open triangle in Figure 1). Assays for determining the percentage of bisecting N-glycans) are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of fucosylated N-glycans is the percent of bisecting N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises less than 10%, 8%, 5%, 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycans. In some embodiments, an anti-CD20 antibody composition provided herein comprises between 10% and 0.5% sialylated glycans; between 10% and 5% sialylated glycans; between 5% and 0.5% sialylated glycans; between 4% and 0.5% sialylated glycans; between 2% and 0.5% sialylated glycans; or no detectable amount of sialylated glycans.
  • Assays for determining the percentage of sialylation are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of sialylated N-glycans is the percent of sialylated N- glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises between at least no detectable amount, 0.5%, 1%, 2%, 3%, 4%, or at least 5% and at most 0.5%, 1%, 2%, 3%, 4%, 5% or at most 10% sialylated glycans. In some embodiments, an anti-CD20 antibody composition provided herein comprises no detectable amount of sialylated glycans.
  • an anti-CD20 antibody composition provided herein comprises between 5% and 15% GOB N-glycans; between 9% and 11% GOB N-glycan; or about 10% GOB N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • GOB N- glycans are shown in Figure 1. Assays for determining the percentage of GOB N-glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of GOB N-glycans is the percent of GOB N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises between at least 5%, 6%, 7%, 8%, 9%, 10%, or at least 11% and at most 7%, 8%, 9%, 10%, 11%, 12% or at most 13% GOB N-glycans.
  • an anti-CD20 antibody composition provided herein comprises between 0.1% and 1.5% Man5 N-glycans; between 0.4% and 0.7% GOB N-glycan; or about 0.6% Man5 N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • Man5 N-glycans are shown in Figure 1. Assays for determining the percentage of Man5 N-glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of Man5 N-glycans is the percent of Man5 N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises between at least 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, or at least 0.7% and at most 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, or at most 0.9% Man5 N-glycans.
  • Man5 N-gylcan is the only high mannose species in the N- glycan profile.
  • an anti-CD20 antibody composition provided herein comprises between about 42% and about 52.8% GO N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • GO N-glycans are shown in Figure 1.
  • Assays for determining the percentage of GO N-glycans are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of GO N-glycans is the percent of GO N-glycans among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided herein comprises between at least 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47% or at least 48% and at most 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 52.8%, 53%, 54%, 55%, 56%, 57% or at most 58% GO N-glycans.
  • an anti-CD20 antibody composition provided is characterized by a specified abundance ratio of G1 to GO N-glycans.
  • the abundance ratio of G1 to GO N- glycans can be between 0.02 and 0.3; between 0.05 and 0.25; between 0.08 and 0.22; between 0.09 and 0.2; between 0.1 and 0.19; between 0.1 and 0.18; between 0.1 and 0.17; between 0.1 and 0.16; or the abundance ratio of G1 to GO N-glycans can be between 0.1 and 0.15.
  • G1 and G1 N-glycans are shown in Figure 1. Assays for determining the percentage of G1 or GO glycans are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of G1 or GO glycans is the percent of G1 or GO glycans, respectively, among N-glycans that were cleaved using the enzymatic digest.
  • an anti-CD20 antibody composition provided is characterized by a specified abundance ratio of GIF to G1 N-glycans.
  • the abundance ratio of GIF to G1 N- glycans can be between 0.1 and 1.2; between 0.2 and 1.1; between 0.3 and 1; between 0.4 and 1; between 0.5 and 1; or the abundance ratio of G1 to GO N-glycans can be between 0.5 and 0.9.
  • GIF and G1 N-glycans are shown in Figure 1. Assays for determining the percentage of GIF or G1 glycans are described in Section 7.5.
  • a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core.
  • the resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry.
  • the percent of GIF or G1 glycans is the percent of GIF or G1 glycans, respectively, among N-glycans that were cleaved using the enzymatic digest.
  • the population of anti-CD20 antibody proteins as described herein comprises asparagine deamidation at one or more asparagine residues present in a heavy chain.
  • the one or more deamidated asparagine residues present in the heavy chain are selected from Asn-33 (as shown in SEQ ID NO: 18), Asn-55 (as shown in SEQ ID NO:19), Asn-61 (as shown in SEQ ID NO: 19), Asn-160 (as shown in SEQ ID NO:22), Asn-202 (as shown in SEQ ID NO:22), Asn-204 (as shown in SEQ ID NO:22), Asn-277 (as shown in SEQ ID NO:25), Asn-287 (as shown in SEQ ID NO:25), Asn-362 (as shown in SEQ ID NO:27), or Asn-385 (as shown in SEQ ID NO:28).
  • the population of anti-CD20 antibody proteins as described herein comprises asparagine deamidation at one or more asparagine residues present in a light chain.
  • the one or more deamidated asparagine residues present in the light chain are selected from Asn-136 (as shown in SEQ ID NO:31), Asn-137 (as shown in SEQ ID NO:31), Asn-151 (as shown in SEQ ID NO:32), or Asn-157 (as shown in SEQ ID NO:32).
  • the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a heavy chain.
  • the one or more methionine residues present in the heavy chain are selected from Met-20, Met-34, Met-81, Met-253 or Met-428 as shown in SEQ ID NO: 17, 18, 21, 24, or 29, respectively.
  • the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a light chain.
  • the one or more methionine residues present in the light chain are selected from Met-21 or Met-32 as shown in SEQ ID NO:30.
  • the population of anti-CD20 antibody proteins comprises pyroglutamation at the N-terminal glutamine residue present in a heavy chain or a light chain.
  • the pyroglutamation at the N-terminal glutamine residue is present in the heavy chain, for example, pGlu-1 as shown in SEQ ID NO: 13.
  • the pyroglutamation at the N-terminal glutamine residue is present in the light chain, for example, pGlu-1 as shown in SEQ ID NO: 14.
  • the glutamate at position 1 of the heavy chain is a pyroglutamate and the glutamate at position 1 of the light chain is a pyroglutamate.
  • the population of anti-CD20 antibody proteins comprises a deletion of the C-terminal lysine amino acid residue present in a heavy chain or a light chain. In some embodiments, the C-terminal lysine amino acid residue of the heavy chain in the anti- CD20 antibody in said population is truncated. In some embodiments, the population of anti- CD20 antibody proteins comprises a deletion of the C-terminal lysine at the heavy chain. In some embodiments, the heavy chain of the anti-CD20 antibody proteins comprises the amino acid sequence of SEQ ID NO: 15.
  • compositions or population of anti-CD20 antibody proteins provided herein e.g., such that are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2 have a specified three-dimensional folding pattern or confirmation.
  • the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at a range of 3.0% to 15.0%; Anti-parallel P-sheet at a range of 25.0% to 40.0%; Parallel P-sheet at a range of 0.5% to 12.0%; P-Turn at a range of 10.0% to 25.0%; and random coil at a range of 30.0% to 42.0%.
  • CD circular dichroism
  • the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at a range of 8.0% to 10.0%; Anti-parallel P-sheet at a range of 32.0% to 36.0%; Parallel P-sheet at a range of 5.0% to 6.0%; P-Turn at a range of 16.0% to 18.0%; and random coil at a range of 35.0% to 36.0%.
  • CD circular dichroism
  • the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at about 9.0%; Anti-parallel P-sheet at about 33.0%; Parallel P-sheet at about 5.6%; P-Turn at about 17.5%; and random coil at about 35.2%, wherein the term “about” means ⁇ 5%.
  • CD circular dichroism
  • provided herein is a method of determining the amount of post-translational modifications in a population of anti-CD20 antibody proteins.
  • the post-translational modification is selected from asparagine deamidation, methionine oxidation, glycosylation, pyroglutamate formation, and lysine truncation.
  • the method of determining the amount of post-translational modifications in a population of anti-CD20 antibody proteins comprises a step of digesting the population of anti-CD20 antibody proteins with an endoproteinase.
  • the population of anti-CD20 antibody proteins is reduced prior to digestion.
  • the population of anti-CD20 antibody proteins is alkylated prior to digestion.
  • the endoproteinase is selected from Asp-N, Lys-C, or trypsin.
  • the step of digestion occurs at 37°C for at least 8 hours, at least 12 hours, at least 16 hours, at least 20 hours, overnight, and/or less than 24 hours.
  • the population of anti-CD20 antibody proteins is digested using Asp-N or Lys-C at 37°C overnight. In some embodiments, the population of anti-CD20 antibody proteins is digested using trypsin at ratio of 50: 1 (w:w) at 37°C overnight. In some embodiments, the digested population of anti- CD20 antibody proteins is purified from digestion reaction components and/or undigested anti- CD20 antibody proteins.
  • the method comprises a step for deglycosylating a population of anti-CD20 antibody proteins, thereby producing released N-glycans for labeling.
  • deglycosylating comprises breaking the glycosidic bond between one or more or all N-glycans from a population of anti-CD20 antibody proteins.
  • deglycosylating comprises releasing some or most or substantially all of the N-glycans from a population of anti-CD20 antibody proteins.
  • deglycosylating releases greater than 50 percent, greater than 60 percent, greater than 70 percent, greater than 80 percent, greater than 90 percent, greater than 95 percent, greater than 97 percent, greater than 98 percent, greater than 99 percent, or 100 percent of N-glycans present on a population of anti-CD20 antibody proteins.
  • deglycosylating comprises contacting the population of anti- CD20 antibody proteins with one or more deglycosylating reagent, which cleaves N-glycans or N-linked oligosaccharides.
  • the deglycosylating reagent is a deglycosylating enzyme or chemical agent.
  • the deglycosylating enzyme is PNGase F.
  • deglycosylating comprises contacting the population of anti- CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent at a deglycosylation temperature of from about 25°C to about 50°C, from about 37°C to about 50°C, about 25 C, about 37°C, about 42°C, or about 50°C. In some embodiments, the deglycosylation temperature is 37°C. The rate of deglycosylation may be increased by increasing the deglycosylation temperature.
  • deglycosylation comprises contacting a population of anti-CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent for a period of time sufficient to release some or most or substantially all of the N-glycans from the population of anti-CD20 antibody proteins.
  • deglycosylating comprises contacting a population of anti-CD20 antibody proteins with said one or more deglycosylating enzyme or chemical agent for at least 30 minutes, at least 1 hour, at least 2 hours, at least 4 hours, at least 8 hours, at least 12 hours, at least 16 hours, at least 20 hours, at least 24 hours, at least 30 hours, and/or less than 48 hours.
  • deglycosylating comprises contacting a population of anti-CD20 antibody proteins with said one or more deglycosylating enzyme or chemical agent for a period of time of from about 30 minutes to about 2 hours, about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 20 hours, about 20 to about 30 hours, greater than about 30 hours, and/or less than 48 hours.
  • deglycosylating comprises contacting a population of anti-CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent for about 12 to about 20 hours.
  • deglycosylation comprises contacting a population of anti-CD20 antibody proteins with PNGase F at about 37 °C for about 12 to about 20 hours. In some embodiments, deglycosylation comprises contacting an anti- CD20 antibody with PNGase F at 37 °C for a time period of from 12-20 hours. In some embodiments, deglycosylation with PNGase F occurs in the presence of a non-ionic detergent (i.e., NP-40).
  • a non-ionic detergent i.e., NP-40
  • deglycosylation comprises denaturing the population of anti- CD20 antibody proteins prior to contacting the population with said one or more deglycosylating enzyme or chemical agent.
  • denaturing comprises thermal denaturation, chemical denaturation, or a combination of both.
  • thermal denaturation comprises incubating a population of anti-CD20 antibody proteins at a denaturation temperature and denaturation time sufficient to unfold some or most or all of the immunoglobulin fold domains of anti-CD20 antibody proteins of said population.
  • the denaturation temperature is at least 50°C, at least 60 °C, at least 65 °C, at least 70 °C, at least 75 °C, at least 80 °C, at least 85 °C, at least 90 °C, and/or less than about 100 °C. In some embodiments, the denaturation temperature is from about 50 °C to about 60 °C, from about 60 °C to about 70 °C, from about 70 °C to about 80 °C, from about 80 °C to about 90°C, or from 90 °C to about 100 °C.
  • the denaturation temperature is about 50 °C, about 60 °C, about 65 °C, about 70 °C, about 75 °C, about 80 °C, about 85 °C, about 90 °C, or about 100 °C. In some embodiments, the denaturation temperature is 70 °C. In some embodiments, chemical denaturation comprises incubating a population of anti-CD20 antibody proteins at a denaturation temperature of at least 25 °C, at least 30 °C, at least 37 °C, or an elevated temperature (i.e., a thermal denaturation temperature).
  • the denaturation time is at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 4 hours, at least 10 hours, or at least 24 hours. In some embodiments, the denaturation time is about 5 minutes, about 10 minutes, about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, about 4 hours, about 10 hours, or about 24 hours. In some embodiments, the denaturation time is 10 minutes.
  • chemical denaturation comprises contacting the population of anti-CD20 antibody proteins with one or more chemical denaturant.
  • the one or more chemical denaturant is selected from an ionic detergent, non-ionic detergent, zwitterionic detergent, chaotropic detergent, or reducing agent.
  • the chemical denaturant is selected from sodium dodecyl sulfate (SDS), urea, or dithiothreitol (DDT).
  • SDS sodium dodecyl sulfate
  • DDT dithiothreitol
  • the effects of certain chemical denaturants may interfere with deglycosylation (i.e., enzymatic deglycosylation) and can be countered in solution.
  • the chemical denaturant is countered by addition of a non-ionic detergent to the solution.
  • the chemical denaturant is SDS and the additional non-ionic detergent is NP-40.
  • denaturing a population of anti-CD20 antibody proteins comprises thermal denaturation at 70 °C for 10 minutes.
  • the method comprises purifying released N-glycans of a population of anti-CD20 antibody proteins from a deglycosylation reaction mixture.
  • the released N-glycans are substantially pure of deglycosylating reagent and deglycosylated or unreacted anti-CD20 antibodies.
  • the released N- glycans are substantially pure of salts and/or detergents.
  • the released N- glycans are purified by hydrophilic interaction.
  • the released N-glycans are purified by chromatography comprising a hydrophilic stationary phase and reversed-phase eluent.
  • the released N-glycans are purified by hydrophilic interaction liquid chromatography (HILIC). In some embodiments, the released N-glycans are purified via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol. In some embodiments, the released N-glycans are purified to a purity level equivalent to the purity level obtained when purifying via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol.
  • HILIC hydrophilic interaction liquid chromatography
  • the method comprises a step for labeling released N-glycans of a population of anti-CD20 antibody proteins, thereby producing labeled N-glycans for detection.
  • labeling released N-glycans comprises chemical derivatization, for example to provide a detectable charge, ultraviolet activity, or fluorescent characteristic to the released N-glycans.
  • labeling released N-glycans comprises reductive amination, hydrazide labeling, methylation, Michael addition, or permethylation.
  • labeling released N-glycans comprises contacting the released N-glycans with a label selected from 2-aminobenzamide (2- AB), 2-aminobenzoic acid (2-AA), 2-aminopyridine (PA), 2-aminonaphthalene trisulfonic acid (ANTS), or 1 -aminopyrene- 3,6,8-trisulfonic acid (APTS).
  • the label is 2-AB.
  • labeling of released N-glycans by reductive amination comprises the use of a reducing agent.
  • the reducing agent is selected from sodium cyanoborohydride or 2-picoline borane.
  • labeling released N-glycans by reductive amination comprises contacting the released N-glycans with a label suitable for reductive amination and a reducing agent, at a reaction temperature and reaction time sufficient for labeling to occur.
  • the reaction temperature is of from about 25 °C to about 40 °C, from about 40 °C to about 50 °C, from about 50 °C to about 60 °C, from about 60 °C to about 70 °C, or from about 70°C to about 80 °C.
  • the reaction temperature is about 25 °C, about 30 °C, about 35 °C, about 40 °C, about 45 °C, about 50 °C, about 55 °C, about 60 °C, about 65 °C, about 70 °C, or about 75°C. In some embodiments, the reaction temperature is 65 °C. In some embodiments, the reaction time is at least 30 minutes, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, or at least 6 hours. In some embodiments, the reaction time is of from about 30 minutes to about 1 hour, from about 1 hours to about 2 hours, from about 2 hours to about 3 hours, from about 3 hours to about 4 hours, or from about 4 hours to about 6 hours.
  • the reaction time is about 30 minutes, about 1 hours, about 2 hours, about 3 hours, about 4 hours, or about 6 hours. In some embodiments, the reaction time is 3 hours.
  • labeling released N- glycans from a population of anti-CD20 antibody proteins comprises reductive amination using 2- AB and cyanoborohydride at 65 °C for 3 hours, thereby producing labeled N-glycans.
  • the method comprises purifying labeled N-glycans of a population of anti-CD20 antibody proteins from a labeling reaction mixture.
  • the labeled N-glycans are substantially pure of unreacted label.
  • the labeled N-glycans are substantially pure of unreacted released N-glycans.
  • the labeled N-glycans are substantially pure of reducing agent (i.e., cyanoborohydride).
  • the labeled N-glycans are purified by hydrophilic interaction.
  • the labeled N-glycans are purified by chromatography comprising a hydrophilic stationary phase and reversed-phase eluent. In certain embodiments, the labeled N-glycans are purified by hydrophilic interaction liquid chromatography (HILIC). In some embodiments, the labeled N-glycans are purified via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol. In some embodiments, the labeled N-glycans are purified to a purity level equivalent to the purity level obtained when purifying via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol.
  • HILIC hydrophilic interaction liquid chromatography
  • the method comprises a step for separating labeled N-glycans of a population of anti-CD20 antibody proteins and determining an N-glycan profile of the population.
  • separating labeled N-glycans comprises separation by hydrophilicity.
  • separating labeled N-glycans comprises chromatography, which comprises a hydrophilic stationary phase and a reversed-phase eluent.
  • the chromatography is hydrophilic interaction liquid chromatography (HILIC).
  • separating labeled N-glycans comprises chromatography comprising an amide stationary phase.
  • the amide stationary phase is a glycan BEH amide column.
  • the reversed-phase eluent comprises one or more mobile phase, wherein the mobile phase comprises an acidic ammonium buffer and/or acetonitrile.
  • chromatography is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate labeled N-glycans.
  • chromatography is performed using a chromatography system that controls these parameters (i.e., an HPLC or UPLC system).
  • the chromatography system is coupled to a detector, for detecting labeled N-glycans of a population of anti-CD20 antibody proteins.
  • the detector is a fluorescence detector that detects fluorescence from the label (e.g., the label that is contacted with the released N-glycans of a population of anti-CD20 antibody proteins in a labeling step).
  • the label is 2-AB.
  • detecting fluorescence comprises exciting with 360 nm wavelength light and observing fluorescent emission at 428 nm wavelength light.
  • the chromatography system is a HILIC-UPLC system.
  • separation and detection of labeled N-glycans is performed using a hydrophilic interaction stationary phase, a revers ed-phase eluent, and a Waters UPLC equipped with fluorescence detector.
  • the hydrophilic interaction stationary phase comprises a glycan BEH amide column (130A, 1.7 pm, 2.1 mm X 150 mm).
  • the stationary phase (i.e., column) temperature is 50 °C.
  • the flow rate is 0.50 mL/min.
  • the reversed-phase eluent comprises one or more mobile phases comprising a first mobile phase comprising about 250 mM ammonium formate, at a pH of about 4.4, and a second mobile phase comprising acetonitrile.
  • the mobile phase gradient comprises an increase of said first mobile phase of from 22.0% to 44.1% over 38.5 minutes.
  • detecting fluorescence comprises generating a chromatogram, wherein the dependent variable is selected from mobile phase volume, eluent volume passed through the chromatographic column, or time, and the observable is fluorescence.
  • determining an N-glycan profile of a population of anti-CD20 antibody proteins comprises quantifying the relative amounts of labeled G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans of said population.
  • chromatography i.e., HILIC-UPLC
  • a flow rate, column temperature, mobile phase gradient, and period of time parameters sufficient to separate the N-glycans of G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F for quantification.
  • the amount of an N- glycan is quantified by calculating its area under a curve comprising labeled G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans in a chromatograph separating said N-glycans.
  • the relative abundance of an N-glycan selected from G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, or G2F is quantified by calculating the percent peak area of said N- glycan relative to total peak area of GO-GN, GOF-GN, GO, Gl-GN, GOB, GOF, Man5, GOFB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans in a chromatograph separating said N- glycans.
  • the peak area of an N-glycan in the chromatograph is greater than or equal to 0.25%. In certain embodiments, the peak area of an N-glycan in the chromatograph has a signal-to-noise ratio of greater than or equal to 3.0.
  • An N-glycan profile of a population of anti-CD20 antibody proteins may be determining the relative amounts of glycosylation of the population via a liquid chromatography- coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific glycosylation results of a digested population of anti-CD20 antibody proteins.
  • LC-MS liquid chromatography- coupled mass spectrometer
  • the LC-MS peptide mapping comprises determining the molecular weight of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of glycosylated residues (i.e., GO, GOF, GOB, GOFB, Gl, or GIF).
  • the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion.
  • the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest.
  • the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
  • the observed molecular weights are compared to theoretical masses for glycosylated peptides selected from Table 1, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 1, column 4. Table 1: Peptides for determining glycosylation following Lys-C digestion [00255] In some embodiments, the population of anti-CD20 antibody proteins comprises glycosylation at residue Asn-298 of SEQ ID NO: 33 present in a heavy chain.
  • Also provided herein is a method of determining the amount of asparagine deamidation in a population of anti-CD20 antibody proteins.
  • the amount of asparagine deamidation in a population of anti-CD20 antibody proteins is the amount of isoaspartate residues in the population.
  • the method comprises detecting the amount of isoaspartate residues in a digested population of anti-CD20 antibody proteins.
  • the amount of asparagine deamidation or isoaspartate in a population or digested population of anti-CD20 antibody proteins may determined via enzymatic method, for example, by the addition of a methyltransferase (i.e., protein isoaspartyl methyltransferase (PIMT)), which catalyzes the transfer of a methyl group from S-adenosyl methionine (SAM) to isoaspartic acid.
  • a methyltransferase i.e., protein isoaspartyl methyltransferase (PIMT)
  • SAM S-adenosyl methionine
  • SAH S-adenosyl homocysteine
  • UPLC ultra performance liquid chromatography
  • RP-HPLC reversed-phase high performance liquid chromatography
  • the method comprises a step of derivatization comprising contacting the population of anti-CD20 antibody proteins with PIMT and SAM.
  • the population of anti-CD20 antibody proteins is contacted with PIMT and SAM at a temperature and for a period of time sufficient for derivatization (i.e., methylation) of substantially all of the aspartate residues present in the population.
  • the method optionally comprises a step of quenching the derivatization reaction.
  • the method comprises a step of separating SAH by hydrophobicity for quantification of isoaspartate.
  • separating SAH comprises chromatography, which comprises a hydrophobic interaction stationary phase and a reversed- phase eluent.
  • separating SAH comprises chromatography using a Cl 8 stationary phase.
  • the Cl 8 stationary phase is a Hydro-RP column.
  • the revers ed-phase eluent comprises one or more mobile phase (i.e., a weakly acidic phosphate buffer and/or methanol).
  • chromatography is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate SAH from the derivatization reaction components.
  • chromatography is performed using a chromatography system that controls these parameters (i.e., an HPLC or UPLC system).
  • the chromatography system is coupled to a detector, for detecting SAH after derivatization (i.e., absorbance at 260 nm).
  • separation and detection of SAM is performed using a hydrophilic interaction stationary phase, a reversed-phase eluent, and a RP-HPLC or UPLC system equipped with detector for absorbance of SAH at about 260 nm.
  • the hydrophobic interaction stationary phase comprises a Synergi Hydro-RP (4.6 mm X 150 mm).
  • the stationary phase (i.e., column) temperature is about 25 °C or room temperature.
  • the flow rate is 1 mL/min.
  • the reversed-phase eluent comprises one or more mobile phases comprising a first mobile phase comprising about 50 mM potassium phosphate, at a pH of about 6.2, and a second mobile phase comprising methanol.
  • the mobile phase gradient comprises said second mobile phase at 10.0% at 0 minutes, 40% at 7.5 minutes, 80% at 10.5 minutes, 80% at 12.5 minutes, 10% at 13.5 minutes, 10% at 20 minutes, and 10% at 25 minutes.
  • the amount of SAH is quantified by comparing its integrated curve area to a SAH standard curve.
  • the SAH standard curve is prepared using Trp-Ala-Gly- Gly-isoAsp-Ala-Ser-Gly-Glu.
  • the amount of asparagine deamidation or isoaspartate in a population of anti-CD20 antibody proteins is determined using the Promega Isoquant kit along with Isoasp-DSIP standard, in accordance to manufacturer’s protocol.
  • the amount of asparagine deamidation or isoaspartate in a population or digested population of anti-CD20 antibody proteins may determined via a liquid chromatography-coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific deamidation results of a digested population of anti-CD20 antibody proteins.
  • the LC-MS peptide mapping comprises determining the molecular weight and/or relative abundance of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of deamidation or isoaspartate residues.
  • the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion.
  • the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest.
  • the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
  • the observed molecular weights are compared to theoretical masses for deamidated peptides selected from Table 2, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 2, column 3. Table 2: Peptides for determining deamidation following Lys-C digestion
  • the LC-MS peptide mapping comprises determining the molecular weight and/or relative abundance of peptides derived from an Asp-N digested population of anti-CD20 antibody proteins to determine the presence and amount of deamidation or isoaspartate residues.
  • the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion.
  • the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Asp-N digest.
  • the relative abundance of a peptide of an Asp-N digest is calculated by integrating its area under the curve compared to total area.
  • the observed molecular weights are compared to theoretical masses for deamidated peptides selected from Table 3, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 3, column 3.
  • Also provided here is a method of determining the level of methionine oxidation in a population of anti-CD20 antibody proteins as described herein.
  • the amount of methionine oxidation in a population of anti-CD20 antibody proteins is the amount of Met sulfoxide (MetO) residues in the population.
  • the method comprises detecting the amount of Met sulfoxide (MetO) residues in the population.
  • the amount of methionine oxidation or Met sulfoxide (MetO) residues in a population or digested population of anti-CD20 antibody proteins may be determined via a liquid chromatography-coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific oxidation results of a digested population of anti-CD20 antibody proteins.
  • LC-MS peptide mapping comprises determining the molecular weight of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of oxidation or MetO residues.
  • the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion.
  • the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest.
  • the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
  • the observed molecular weights are compared to theoretical masses for oxidized peptides selected from Table 4, column 2. In some embodiments, the observed molecular weights are compared to theoretical masses selected from Table 4, column 3.
  • the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a heavy chain.
  • the one or more methionine residues present in the heavy chain are selected from Met-20, Met-34, Met-81, Met-253 or Met-428 as shown in SEQ ID NO: 17, 18, 21, 24, or 29, respectively.
  • the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a light chain.
  • the one or more methionine residues present in the light chain are selected from Met-21 or Met-32 as shown in SEQ ID NO:30.
  • the secondary structures of anti-CD20 antibody proteins are analyzed using Circular Dichroism (CD) spectroscopy by measuring the difference in absorption between left and right circularly polarized light due to structural asymmetry.
  • CD Circular Dichroism
  • the CD spectroscopy using far-ultraviolet spectra with a wavelength between approximately 170 and 260 nm.
  • the different secondary structures commonly found in protein can be analyzed, since a-helix, parallel and anti-parallel
  • compositions or populations of anti-CD20 antibody proteins described in Section 7.4 at scaled up quantities are present in a single batch, ie, a composition derived from a single run from a single bioreactor of a specified volume.
  • the anti-CD20 antibody proteins obtained from a single run of a 15,000L bioreactor can be referred to as a single batch.
  • the bioreactor has a capacity of at least 100; 200; 300; 400; 500; 750; 1,000; 2,000; 3,000; 4,000; 5,000; 7,500; 10,000; 15,000; 20,000; or at least 25,000 L.
  • anti-CD20 antibody proteins are present in such a single batch at a concentration of at least 10 mg/ml;
  • anti-CD20 antibody proteins are present in such a single batch at a concentration of at between 10 to 35 mg/ml; 10 to 30 mg/ml; 10 to 25 mg/ml; 10 to 20 mg/ml; 10 to 15 mg/ml; 15 to 35 mg/ml; 15 to 30 mg/ml; 15 to 25 mg/ml; 15 to 20 mg/ml; 20 to 35 mg/ml; 20 to 30 mg/ml; 20 to 25 mg/ml; 25 to 35 mg/ml; or 25 to 30 mg/ml as determined using an assay described in Section 7.6(a).
  • anti-CD20 antibody proteins are present in such a single batch at a concentration of about 15mg/ml; about 20mg/ml; about about 25mg/ml; about 30mg/ml; or about 35mg/ml as determined using an assay described in Section 7.6(a) (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
  • the anti-CD20 antibody proteins disclosed herein are produced at a commercial scale.
  • the commercial scale is 10,000 to 25,000 L.
  • the amount of total protein is measured using spectrophotometry. In certain embodiments, the amount of total protein is measured by absorbance at 280 nm.
  • the amount of total protein can be measured using the following assay procedures.
  • This method determines the concentration of the anti-CD20 antibody proteins using an ultraviolet absorbance measurement at 280 nm with a light scattering correction at 320 nm. The calculation of concentration is based on the Beer-Lambert law.
  • the extinction coefficient of the anti-CD20 antibody protein at 280 nm is 1.61 mL/mg/cm as determined by the amino acid composition. The extinction coefficient can be used.
  • the method is suitable for use for post Protein A purification in-process sample testing, release testing, and stability testing. Test samples are diluted gravimetrically in triplicate with 0.9% sodium chloride to a target protein concentration of 0.4 mg/mL.
  • Bovine serum albumin is used as a system suitability control and is tested before and after sample measurements. Test samples are loaded into a quartz cuvette with a 1 cm pathlength and the absorbances at 280 and 320 nm are measured. The test samples are tested in triplicate. In certain embodiments, the amount of total protein can be measured using the critical material and equipment listed in Table 5. In certain embodiments, the amount of total protein can be measured using the protein concentration system suitability criteria listed in Table 6.
  • the population of anti-CD20 antibody proteins provided herein has an amount of total protein of 25.5-25.8 mg/mL. In certain embodiments, the population has an amount of total protein of about 25.6 mg/mL.
  • the populations of anti-CD20 antibody proteins provided herein possess biological properties detailed in this section.
  • the biological properties can be measured using the assays described in Section 7.7(a).
  • the biological properties of the anti-CD20 antibody protein compositions provided herein are described in Section 7.7(b).
  • ADCC Cell-based antibody-dependent cellular cytotoxicity
  • the cell-based ADCC assay uses Raji cells as target cells. In certain embodiments, the Raji cells express CD20. In certain embodiments, the cell-based ADCC assay uses effector cells selected from CD 16 effector cells and primary NK cells. In certain embodiments, the cell-based ADCC assay uses Raji cells as target cells and KILR CD16a effector cells effector cells. In certain embodiments, the cell-based ADCC assay uses Raji cells as target cells and primary NK cells as effector cells. In certain embodiments, the KILR CD 16a effector cells are single donor-derived human CD 8+ T-lymphocytes engineered to express CD16/FcyRIII on their plasma membrane surface.
  • the cell-based ADCC assay uses target cell lysis as the read out.
  • the target cell lysis is quantified using CytoTox GioTM (Promega).
  • the cell-based ADCC assay shows relative potency of the population against a commercial reference standard.
  • the cell-based ADCC assay produces a dose-response curve and an EC50 value.
  • a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells at more than 100% of that of commercial reference standard RS-117808.
  • the ADCC activity can be measured using a CD 16 activity assay.
  • the CD 16 activity assay assesses ADCC activity using a surrogate read-out.
  • the CD 16 activity assay uses WIL2-S as target cells.
  • the effector cells used are a stably transformed Jurkat cell line expressing a chimeric molecule comprising of the extracellular domain of FcyRIIIa joined to the transmembrane and intracellular domains of the gamma chain of the mast cell/basophil Fc receptor for IgE.
  • cells are combined and treated with a serial dilution of the anti-CD20 antibody in the presence of PMA (Phorbol 12- Myristate 13-Acetate).
  • the activation of the effector cells induces the release of IL-2, which is measured by a commercial ELISA kit.
  • the potency is reported as a percentage relative to a reference standard.
  • the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard.
  • the population has a relative potency of about 115% in a CD 16 activity assay compared to a commercial reference standard.
  • the commercial reference standard is RS- 117808.
  • the cell-based ADCC assay uses the following assay procedures. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are used in the concentration range of 250.00 pg/mL to 0.04 pg/mL (250, 50, 16.7, 5.6, 1.9, 0.6, 0.2, 0.04 pg/mL). Two independent preparations of the materials are prepared and assayed across duplicate plates. Assay controls are prepared in triplicates and include: Target Cells Alone Control, Target Cells Death Control, Effector Cells Alone Control, and Effector & Target Cells Control. KILR cells are obtained from Eurofins. They come from a master cell bank made from a unique human donor.
  • a Master and Working cell bank system and qualification protocol allows the vendor to create many thousands of vials in a reproducible manner and TG maintains a secure supply of this critical reagent. Procedures, such as incoming material qualification process, provide assurance that highly reproducible cells are used in every assay.
  • KILR cells are thawed and cultured at 1x10 A 6 cells/mL in media supplemented with IL-2, and are rested for a minimum of 6 days before use and are not be used beyond 14 days post thaw.
  • KILR ADCC To perform the KILR ADCC assay, Raji cells, also managed through a master and working cell banking system, are seeded at 1x10 5 cells/mL, and reference standard, internal assay control, and testing samples dilutions are added to the Raji cells.
  • KILR Effector cells are seeded at 5x10 5 cells/mL to a final effector: target (E:T) ratio of 5: 1.
  • E:T effector: target
  • Cells are cultured at 36 ⁇ 1°C, 5 ⁇ 1%CO2 for 18 to 22 hours.
  • a CytoTox GLoTM preparation is added, and plates are incubated for 30 ⁇ 10 minutes. The plates are read using the SpectraMax plate reader.
  • SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data is evaluated using the SoftMax Pro software for potency against the reference standard. Representative reference standard and sample dose response curves are shown. Results are reported as percentage potency relative to the primary reference standard or its derivative.
  • the cell-based ADCC assay is used as a potency assay for batch release. In some embodiments, the cell-based ADCC assay is used as a potency assay for stability testing. In some embodiments, the cell-based ADCC assay is used as a potency assay in the manufacturing quality controls and processes. In some embodiments, the cell-based ADCC assay is used in comparison studies (for example, in research and development or clinical studies).
  • the higher potency of the population in a cell-based ADCC assay is related to the lower level of fucose content in the N-glycan profile of the population. In some embodiments, the higher potency of the population in a cell-based ADCC assay is due to the lower level of fucose content in the N-glycan profile of the population.
  • ADCP Antibody-dependent cellular phagocytosis
  • the antibody-dependent cellular phagocytosis (ADCP) activity can be measured using CD20 expressing Daudi cells as target cells (labeled by PKH26).
  • human monocytes are isolated from PBMC and differentiated in vitro using GM- CSF to yield macrophages.
  • macrophages are co-cultured with PKH26- labeled target cells previously incubated with serial diluted anti-CD20 antibody samples.
  • target cell phagocytosis was assessed by flow cytometry.
  • the dose response curves, as well as calculated IC50s of the samples tested can be shown.
  • the cell-based CDC assay uses Jeko-1 cells as target cells and rabbit compliment system. In certain embodiments, the cell-based CDC assay uses Raji cells as target cells and human complement system. In some embodiments, the cell-based CDC assay uses target cell lysis as the read out.
  • the cell-based CDC assay uses the following assay procedures.
  • Nine-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are used in the concentration range of 10,000 ng/mL to 10.42 ng/mL (10,000.00, 3333.33, 1666.67, 833.33, 416.67, 208.33, 104.17, 52.08, 10.42 ng/mL).
  • Two independent preparations of the materials are prepared and assayed across duplicate plates.
  • Assay negative controls are prepared in triplicates and include target cells and complement control and target cells alone control.
  • Jeko-1 cells obtained from the ATCC and maintained through a master banking system, are seeded at 3x10 5 cells/mL and incubated for 60-90 minutes. Reference standard, internal assay control, and test samples diluted in duplicate in cell culture medium are added. Complement is then added to the wells and the plates are incubated for approximately 2 hours at 36°C ⁇ 1 °C followed by 25 ⁇ 5 minutes at room temperature. Controls include a target cells with complement only control and a target cell only control to provide a basal level of target cell viability over the course of the assay. The Cell Titer-Gio reagent is then added and incubated an additional 30 ⁇ 10 minutes at room temperature.
  • the plates are read using a SpectraMax M5 plate reader.
  • SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data are evaluated using the Softmax Pro software for potency against the reference standard. Results are reported as % potency relative to the reference standard. Representative reference standard and sample dose response curves are shown.
  • the cell-based CD20 binding assay uses Jeko-1 cells and an MSD assays. In some embodiments, the cell-based CD20 binding assay produces a dose dependent binding curve and a EC50 value.
  • the cell-based CD20 binding assay uses the following assay procedures. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are prepared in the concentration range of 40,000.00 ug/mL to 0.23 ng/mL (40,000.00, 4,000.00, 1,000.00, 333.30, 111.10, 37.00, 4.60, 0.23 ng/mL). Two independent preparations of the Test Material are prepared for each 2-plate Test Material assessment. Assay controls include No cell control (Reference Standard/Test Material dilution + detection reagent, omitting cells) and Cell only control (Cells + detection reagent, omitting Reference Standard/Test Material).
  • Jeko-1 cells are seeded onto MSD high bind plate in PBS at 3 x 10 5 cells per mL, in a final volume of 100 pL per well, and incubated at 35 to 37°C for 2 hours ⁇ 10 minutes. Unbound cells are removed by a PBS wash, the plates are blocked with 45% FBS. Fifty pL of Reference Standard, QC Reference Control, or Test Materials dilutions are added, and the plate is incubated at room temperature for 1 hour ⁇ 10 min while shaking. Following incubation and three PBS washes, 50 pL anti-human Fc detection antibody conjugated with STREP- SULFOTAG is added and incubated for 1 hour ⁇ 5 minutes at room temperature while shaking.
  • the plates are washed again with PBS and 150 pL of the MesoScale read buffer, containing tripropylamine (TP A), is added as a co-reactant for light generation for an electrochemiluminescence read out. Plates are read immediately on a MSD Reader using Workbench 4.0. The resulting data are evaluated using the PLA software and analyzed using a constrained 4 parameter logistic model to generate a relative binding, 95% confidence intervals and results relative to the reference standard. Binding activity results are reported as percentage potency relative to the reference standard. Representative reference standard and testing sample dose response curves, from which relative potency test result is calculated.
  • TP A tripropylamine
  • the FcyRIIIa binding assay uses Surface Plasmon Resonance (SPR).
  • the FcyRIIIa binding assay produces sensorgrams showing a dose dependent binding, saturation, and dissociation.
  • the FcyRIIIa binding assay calculates dissociation constant by the on and off rate and steady state kinetics.
  • the binding affinity to FcyRIIIa 158V of the population is approximately one order of magnitude higher than binding affinity to FcyRIIIa 158F of the population.
  • the binding affinity to FcyRIIIa 158V of the population is significantly higher than Rituxan.
  • the binding affinity to FcyRIIIa 158F of the population is significantly higher than Rituxan. In certain embodiments, the binding affinities of the population to both FcyRIIIa 158V and FcyRIIIa 158F are significantly higher than Rituxan.
  • the FcyRIIIa binding assay uses the following assay procedure. FcyRIIIa 158V receptor (1.2 pg/ml) is immobilized on the chip surface using covalent amine coupling chemistry. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are prepared in the concentration range of 2000 nM to 15.6 nM with a dilution factor of 2.
  • KD is determined using steady state affinity for the FcyRIIIa 158F variant.
  • the binding signals are exported into PLA to determine the relative binding response. Results are reported as % potency relative to the reference standard for each FcyRIIIa variant, 158V and 158F.
  • the Clq binding activity can be as measured using ELISA. In certain embodiments, the Clq binding activity can be as measured using the following assay procedure. A 7-point dilution series of the reference standard, QC reference control, and test materials are prepared in the concentration range of 15.00 pg/mL to 0.12 pg/mL. Reference standard, QC reference control, and test materials dilutions are coated onto an ELISA plates, and the plates incubated for 1 hour ⁇ 30 minutes at room temperature (shaking 150 to 200 rpm).
  • the plates are washed (3x with PBS/0.05% Tween), blocked (with 1% BSA and incubated for 1 hour ⁇ 10 minutes at room temperature and shaking 150 to 200 rpm), and washed (3x with PBS/0.05% Tween).
  • Clq that has been conjugated with peroxidase is then added, and the plates incubated for 1.5 hours ⁇ 30 minutes at room temperature (shaking 150 to 200 rpm).
  • a tetramethylbenzidine (TMB) substrate solution is added and plates incubated at room temperature for 7 minutes (-1 minute/ ⁇ 30 seconds). This produces a colorimetric reaction which is proportional to the level of Clq bound.
  • TMB tetramethylbenzidine
  • the reaction is stopped with the addition of IM sulfuric acid and the color is measured at 450 nm using a Molecular Devices SpectraMax microplate reader.
  • SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data are evaluated using the SoftMax Pro software for potency against the reference standard. Clq binding activity results are reported as percentage potency relative to the reference standard. Representative dose response curves from one assay can be shown.
  • the B-cell depletion activity can be measured in a human whole blood B-cell depletion assay. In certain embodiments, the B-cell depletion activity can be measured in an autologous normal human whole blood B-cell depletion assay. In certain embodiments, the B-cell depletion can be measured by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3 -positive T cells, CD 19-positive B-cells, and CD20- positive B-cells. In certain embodiments, percent of B-cell depletion (100-([100/B-/T-cell ratio in sample without antibody] x [B-/T-cell ratio in sample containing antibody])) can be calculated and plotted against sample concentration.
  • the B-cell depletion activity can be measured using blood from three healthy donor(s).
  • the B-cell depletion can be measured by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3-positive T cells, CD 19-positive B-cells, and CD20-positive B-cells.
  • the biological properties of a composition or population of anti-CD20 antibody proteins provided herein can be measured and described in an assay described in Section 7.7(a) and with the use of a comparison with a reference standard.
  • the reference standard is a commercial reference standard.
  • the reference standard is an anti-CD20 antibody.
  • the reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the commercial reference standard is RS-117808.
  • the populations of anti-CD20 antibody proteins provided herein possess the biological properties as shown in Table 7.
  • RS-117808 (“antibody Ublituximab (TG-1101)”) was deposited according to the terms of the Budapest Treaty at the American Type Culture Collection (ATCC), located at 10801 University Boulevard, Manassas, VA 20110, received by the ATCC on April 15, 2022, and assigned unofficial patent deposit number PTA-127294.
  • a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells assay (see Section 7.7(a)(i)) with are relevant potency that is at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, or at at least 200% of that of the commercial reference standard RS-117808.
  • a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells assay (see Section 7.7(a)(i)) with are relevant potency that is at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150- fold, or at least 200-fold of that of the commercially available Rituxan® (Genentech/Biogen).
  • the population induces cytotoxicity in a cell-based antibodydependent cellular cytotoxicity (ADCC) assay.
  • ADCC antibodydependent cellular cytotoxicity
  • the population induces greater cytotoxicity in an ADCC assay compared to a commercial reference standard.
  • the population induces greater cytotoxicity in an ADCC assay compared to an anti-CD20 antibody. In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to GAZYVA (obinutuzumab).
  • the population induces greater cytotoxicity in an ADCC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in a cell-based ADCC assay compared to a commercial reference standard.
  • the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a cell-based ADCC assay compared to a commercial reference standard.
  • the population has a relative potency of at least more than 100% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a cell-based ADCC assay compared to a commercial reference standard.
  • the population has a relative potency of between at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50- fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a cell-based ADCC assay compared to a commercial reference standard.
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300- fold in a cell-based ADCC assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative potency of about 38% to about 3 -fold in a cell-based ADCC assay compared to GAZYVA (obinutuzumab).
  • the population has a relative potency of about 10-fold to about 31 -fold in a cell-based ADCC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 28-fold to about 2250-fold in a cell-based ADCC assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 25-fold in a cell-based ADCC assay compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of 60 to 200%, 70 to 190%, 80% to 180%, 85 to 170%, or 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of about 117% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
  • the EC50 value for the ADCC potency as measured in a cellbased ADCC assay of the population is between 2 and 6 pg/mL. In certain embodiments, the EC50 value for the ADCC potency as measured in a cell-based ADCC assay of the population is between 0.2 and 20 pg/mL, 0.3 and 18 pg/mL, 0.4 and 15 pg/mL, 0.5 and 12 pg/mL, 0.6 and 10 pg/mL, 0.7 and 9 pg/mL, 0.8 and 8 pg/mL, 0.9 and 7 pg/mL, or 1 and 6 pg/mL.
  • the EC50 value for the ADCC potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty or more repeated cell-based ADCC assays.
  • the EC50 value for the ADCC potency as measured in a cell-based ADCC assay e.g., using the Eurofins/DiscoverX KILR CD16a effector cells of the population is about 5.45 pg/mL.
  • the EC50 value for the ADCC potency as measured in a cell-based ADCC assay is about 2.42 pg/mL.
  • ADCP Antibody-dependent cellular phagocytosis
  • the population induces antibody-dependent cellular phagocytosis (ADCP). In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to an anti-CD20 antibody.
  • ADCP antibody-dependent cellular phagocytosis
  • the population induces greater phagocytosis in an ADCP assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • the population induces phagocytosis in an ADCP assay compared to GAZYVA (obinutuzumab).
  • the population induces greater cytotoxicity in a CDC assay compared to ARZERRA (ofatumumab).
  • the population induces greater phagocytosis in an ADCP assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in an ADCP assay compared to a commercial reference standard.
  • the population has a relative potency of between at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at most 300-fold in an ADCP assay compared to a commercial reference standard.
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100- fold, 100-fold and 150-fold, or between 150-fold and 300-fold in an ADCP assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative potency of about 7-fold in an ADCP assay compared to ARZERRA (ofatumumab).
  • the population has a relative potency of about 15-fold in an ADCP assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 23 -fold in an ADCP assay compared to GAZYVA (obinutuzumab).
  • the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 0.1 and 1 ng/mL. In certain embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 1 and 10, 2 and 9, 3 and 8, 4 and 7, or 5 and 6 ng/mL.
  • the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 0.05 and 20 ng/mL, 0.1 and 19 ng/mL, 0.15 and 18 ng/mL, 0.2 and 18 ng/mL, 0.25 and 15 ng/mL, 0.3 and 12 ng/mL, 0.3 and 10 ng/mL, 0.3 and 9 ng/mL, 0.3 and 8 ng/mL, 0.3 and 7 ng/mL, or 0.3 and 6 ng/mL.
  • the EC50 value for the ADCP potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated ADCP assays. In certain embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is about 5.50 ng/mL.
  • the population induces complement dependent cytotoxicity (CDC). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to a commercial reference standard. In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to an anti-CD20 antibody.
  • CDC complement dependent cytotoxicity
  • the population induces greater cytotoxicity in a CDC assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • the population induces greater cytotoxicity in a CDC assay compared to GAZYVA (obinutuzumab).
  • the population induces greater cytotoxicity in a CDC assay compared to ARZERRA (ofatumumab).
  • the population induces greater cytotoxicity in a CDC assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces greater cytotoxicity in a CDC assay assay compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a cell-based CDC assay compared to a commercial reference standard.
  • the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200- fold in a cell-based CDC assay compared to a commercial reference standard.
  • the population has a relative potency of at least more than 100% in a cell-based CDC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a cell-based CDC assay compared to a commercial reference standard.
  • the population has a relative potency of between at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a cell-based CDC assay compared to a commercial reference standard.
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a cell-based CDC assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative potency of about 50% in a cell-based CDC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 37% in a cell-based CDC assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 1.8-fold in a cell-based CDC assay compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of 50 to 150%, 60 to 140%, 70 to 130%, 75 to 120%, or 78 to 116% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 78 to 116% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 73 to 128% or 74 to 127% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of about 91% in a cell-based CDC assay compared to that of a commercial reference standard.
  • the commercial reference standard is RS-117808.
  • the EC50 value for the CDC potency as measured in a cellbased CDC assay of the population is between 0.4 and 0.7 pg/mL or between 0.4 and 0.6 pg/mL.
  • the EC50 value for the CDC potency as measured in a cell-based CDC assay of the population is between 0.05 and 5 pg/mL, 0.1 and 4 pg/mL, 0.15 and 3 pg/mL, 0.2 and 2 pg/mL, 0.25 and 1 pg/mL, 0.3 and 0.9 pg/mL, 0.3 and 0.8 pg/mL, or 0.3 and 0.7 pg/mL.
  • the EC50 value for the CDC potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated cell-based CDC assays.
  • the EC50 value for the CDC potency as measured in a cell-based CDC assay of the population is about 0.5 pg/mL.
  • the population possesses CD20 binding activity in a cell-based CD20 binding assay. In certain embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to a commercial reference standard. [00318] In some embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to an anti-CD20 antibody.
  • the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • GAZYVA bisnutuzumab
  • ARZERRA ofatumumab
  • RITUXAN rituximab
  • veltuzumab veltuzumab
  • ZEVALIN ibritumomab tiuxetan
  • OCREVUS OCREVUS
  • the population possesses greater CD20 binding activity compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater CD20 binding activity compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater CD20 binding activity compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces possesses greater CD20 binding activity compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a CD20 binding assay compared to a commercial reference standard.
  • the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a CD20 binding assay compared to a commercial reference standard.
  • the population has a relative potency of at least more than 100% in a CD20 binding compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a CD20 binding compared to a commercial reference standard.
  • the population has a relative potency of between at least 5%
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a CD20 binding assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative potency of about 1.6-fold to about 5.8-fold in a CD20 binding assay compared to GAZYVA (obinutuzumab).
  • the population has a relative potency of about 81% to about 4.3 -fold in a CD20 binding assay compared to ARZERRA (ofatumumab).
  • the population has a relative potency of about 1.6-fold to about 4.1 -fold in a CD20 binding assay compared to RITUXAN (rituximab).
  • the population has a relative potency of 50 to 150%, 60 to 140%, 70 to 130%, 80 to 120%, 90 to 120%, or 92 to 118% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 92 to 118% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 82 to 138% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of about 109% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
  • the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is between 0.05 and 0.1 pg/mL. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is between 0.01 and 0.5 pg/mL, 0.02 and 0.4 pg/mL, 0.03 and 0.3 pg/mL, 0.04 and 0.2 pg/mL, or 0.05 and 0.1 pg/mL.
  • the EC50 value for the CD20 binding potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated cell-based CD20 binding activity bioassays. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is about 0.093 pg/mL. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is about 0.063 pg/mL.
  • the population possesses FcyRIIIa 158V binding activity in an FcyRIIIa binding assay. In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to an anti-CD20 antibody.
  • the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to veltuzumab (IMMU-106).
  • the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to ZE VALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab).
  • the population has a relative FcyRIIIa 158V binding activity of at least 1000%, 750%, 500%, 250%, 100%, 75%, or at least 50% in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158V binding activity of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158V binding activity of at least more than 100% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative relative FcyRIIIa 158V binding activity of more than 100% in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158V binding activity of at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100- fold, 125-fold, 150-fold, 200-fold or at most 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158V binding activity of at least between 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300- fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative FcyRIIIa 158V binding activity of about 3.8-fold in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab).
  • the population has a relative FcyRIIIa 158V binding activity of about 25.6-fold in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 18.7-fold in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 16-fold in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab).
  • the population possesses FcyRIIIa 158F binding activity in a FcyRIIIa binding assay. In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to an anti-CD20 antibody.
  • the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • GAZYVA bisnutuzumab
  • ARZERRA ofatumumab
  • RITUXAN rituximab
  • veltuzumab veltuzumab
  • ZEVALIN ibritumomab tiuxetan
  • OCREVUS OCREVUS
  • the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to veltuzumab (IMMU-106).
  • the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab).
  • the population has a relative FcyRIIIa 158F binding activity of at least 1000%, 750%, 500%, 250%, 100%, 75%, or at least 50% in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158F binding activity of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158F binding activity of at least more than 100% in a cell-FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative relative FcyRIIIa 158F binding activity of more than 100% in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158F binding activity of at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100- fold, 125-fold, 150-fold, 200-fold or at most 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the population has a relative FcyRIIIa 158F binding activity of at least between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150- fold and 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative FcyRIIIa 158F binding activity of about 2.6-fold in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab).
  • the population has a relative FcyRIIIa 158F binding activity of about 21.8-fold in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 10.2-fold in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 9.9-fold in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab). In certain embodiments, the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab.
  • the population has a relative potency of 20 to 300%, 30 to 250%, 40% to 220%, or 50 to 200% in a cell-based FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 50 to 200% in a cell-based FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 76 to 130% or 82 to 130% in a cell-based FcyRIIIa 158V binding assay compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
  • the population has a KD value 55 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 10 to 100 nM, 15 to 90 nM, 10 to 80 nM, or 30 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 30 to 70 nM in an FcyRIIIa-158V binding assay as measured by surface plasmon resonance.
  • the KD value is the average KD value calculated from the KD values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated FcyRIIIa- 158V binding assays.
  • the population has a KD value about 59 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance.
  • the population has a KD value about 64.1 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance.
  • the population has a KD value 600 to 800 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the population has a KD value 100 to 2000 nM, 200 to 1800 nM, 300 to 1700 nM, 400 to 1600 nM, 500 to 1500 nM, 500 to 1200 nM, 600 to 1000 nM, or 600 to 800 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the KD value is the average KD value calculated from the KD values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated FcyRIIIa- 158V binding assays.
  • the population has a KD value 760 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the population has a KD value 680.3 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
  • the population possesses Clq binding activity as measured by ELISA. In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to a commercial reference standard.
  • the population possesses greater Clq binding activity as measured by ELISA compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, possesses greater Clq binding activity as measured by ELISA compared to GAZYVA (obinutuzumab).
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of at least more than 100% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of between at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the population has a relative potency of about 142% in a Clq binding assay as measured by ELISA compared to ARZERRA (ofatumumab).
  • the population has a relative potency of about 123% in a Clq binding assay as measured by ELISA compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 112% in a Clq binding assay as measured by ELISA compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of 30 to 180%, 40 to 170%, 50 to 160%, 60 to 150%, 70 to 140%, 80 to 130%, 85% to 120%, or 88 to 113% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 88 to 113% or 86 to 117% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 86 to 116% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of about 99% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
  • the EC50 value for the Clq binding activity as measured by ELISA of the population is between 1.5 and 3 pg/mL. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is between 0.2 and 9 pg/mL, 0.3 and 8 pg/mL, 0.4 and 7 pg/mL, 0.5 and 6 pg/mL, 0.6 and 5 pg/mL, 0.7 and 4 pg/mL, 0.8 and 3 pg/mL, 0.9 and 2.9 pg/mL, or 1 and 2.8 pg/mL.
  • the EC50 value for the Clq binding activity is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated ELISA experiments. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is about 1.92 pg/mL. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is about 2.6 pg/mL. (vii) B cell depletion activity
  • the population possesses B cell depletion activity as measured in a human whole blood B cell depletion assay. In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab).
  • GAZYVA bisnutuzumab
  • ARZERRA ofatumumab
  • RITUXAN rituximab
  • veltuzumab veltuzumab
  • ZEVALIN ibritumomab tiuxetan
  • OCREVUS ocrelizumab
  • the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to veltuzumab (IMMU-106).
  • the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to OCREVUS (ocrelizumab).
  • the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125- fold, 150-fold, or at least 200-fold in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the population has a relative potency of at least more than 100% in a human whole blood B cell depletion assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the population has a relative potency of between at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a human whole blood B cell depletion assay compared to a commercial reference standard.
  • the commercial reference standard is an anti-CD20 antibody.
  • the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
  • the relapsing form of multiple sclerosis is selected from a clinically isolated syndrome (“CIS”), relapsing-remitting (“RRMS”), or active secondary progressive disease (“SPMS”).
  • the RMS is a CIS.
  • the RMS is RRMS.
  • the RMS is SPMS.
  • the subject is diagnosed with RMS in accordance to McDonald Criteria (2010), or by another suitable method known by one skilled in the art.
  • the method comprises administering to the subject the anti- CD20 antibody composition as described herein in a multi-dosage regimen. In certain embodiments, the method comprises administering to the subject the anti-CD20 antibody composition as described herein by intravenous infusion.
  • anti-CD20 antibody composition described herein can be administered to the subject in a multi-infusion dosage regimen by intravenous infusion.
  • Anti-CD20 antibody compositions described herein can be administered to the subject by intravenous infusion in a multi-infusion dosage regimen for 48 weeks. Additionally, or in the alternative, the anti-CD20 antibody composition described herein can be administered to the subject by intravenous infusion in a multi-infusion dosage regimen for 96 weeks.
  • the multi-infusion dosage regimen comprises first, second, and subsequent intravenous infusions of ublituximab.
  • “subsequent infusions” of ublituximab is any number of infusions after the second infusion.
  • the intravenous infusion comprises a multi-dosage regimen (e.g., a multi-infusion dosage regimen), comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising
  • the intravenous infusion comprises a multi-dosage regimen (e.g., a multi-infusion dosage regimen), comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody composition at described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of S
  • the method comprises a first administration of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) by intravenous infusion to the subject and then a second administration of the anti-CD20 antibody as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated gly
  • an anti-CD20 antibody composition
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising a prescribed dosage of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) herein at day 1; b) a second infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising a prescribed dosage of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) herein at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-gly
  • the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; and subsequent infusions (e.g., the first subsequent infusion and/or the one or more subsequent infusions) are administered over 1 hour.
  • the method comprises a treatment period of at least 48 weeks. In some embodiments, the method comprises a treatment period of at least 96 weeks.
  • the subject is a human. In some embodiments, the human subject is an adult. In some embodiments, the subject has experienced at least one relapse in the previous year prior to treatment or two relapses in the previous two years prior to treatment. In some embodiments, the subject has or had the presence of a T1 gadolinium (Gd)-enhancing lesion in the previous year prior to treatment with the anti-CD20 antibody composition as described herein. In some embodiments, the subject has an Expanded Disability Status Scale (EDSS) score of from 0 to 5.5 prior to treatment with the anti-CD20 antibody composition as described herein.
  • Gd gadolinium
  • EDSS Expanded Disability Status Scale
  • the subject has not been treated with a nonsteroid therapy for MS in the previous five years prior to treatment with the anti-CD20 antibody composition as described herein.
  • the subject is naive to treatment for MS.
  • the subject is negative for hepatitis B virus (HBV).
  • the subject is negative for hepatitis B virus surface antigen (HBsAg).
  • the subject is negative for anti-hepatitis B virus core antibodies.
  • the subject has not been immunized with a vaccine for at least 2 weeks or at least 4 weeks prior to treatment with the anti-CD20 antibody composition as described herein.
  • the subject is pre-medicated with an amount of a corticosteroid about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein.
  • the corticosteroid is methylprednisone or a corticosteroid bioequivalent thereto.
  • the amount of a corticosteroid is about 100 mg methylprednisone.
  • the corticosteroid is dexamethasone or a corticosteroid bioequivalent thereto.
  • the amount of a corticosteroid is from about 10 to 20 mg dexamethasone.
  • the subject is pre-medicated with an amount of an antipyretic about 30 to about 60 minutes prior to administration of the anti- CD20 antibody composition as described herein.
  • the antipyretic is acetaminophen or an antipyretic bioequivalent thereto.
  • the subject is premedicated with an amout of an antihistamine about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein.
  • the antihistamine is diphenhydramine HC1 or an antihistamine bioequivalent thereof.
  • the amount of an antihistamine is from about 25 to 50 mg diphenhydramine HC1.
  • the subject is pre-medicated with an amount of a corticosteroid (as described above) and an amount of an antihistamine (as described above) about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein.
  • the corticosteroid and/or antihistamine are administered to the subject orally.
  • the method alleviates or delays progression of one or more symptoms of MS in the subject.
  • the method reduces the annualized relapse rate (ARR) in the subject following administration of the anti-CD20 antibody (e.g., ublituximab) composition as described herein.
  • ARR is the total number of relapse for a subject divided by the sum of treatment duration (i.e., it is the ratio of the sum of the subject’s RMS relapse counts divided by the sum of the subject’s treatment duration (in years)).
  • ARR is number of Independent Relapse Adjudication Panel (IRAP)- confirmed relapses for a subject per year.
  • IRAP Independent Relapse Adjudication Panel
  • reduction of ARR is assessed at about 96 weeks after the first infusion.
  • the subject achieves a significantly reduced ARR following administration of the multi-infusion dosage regimen of the anti-CD20 antibody composition described here, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • administration of the anti-CD20 antibody composition described here can significantly reduce ARR with a relative reduction in relapse rate of 59%.
  • treatment with the anti-CD20 antibody composition described here results in an ARR of about 0.100 to about 0.050; such as, about 0.100 to about 0.090, about 0.090 to about 0.080, about 0.080 to about 0.070, about 0.070 to about 0.060, or about 0.060 to about 0.050 (e.g., about 0.099, 0.098, 0.097, 0.096, 0.095, 0.094,
  • the method reduces the total number of T1 gadolinium (Gd)- enhancing lesions in the subject following administration of the anti-CD20 antibody composition as described herein.
  • reduction of Gd-enhancing T1 lesions is assessed by MRI scan.
  • reduction of Gd-enhancing T1 lesions is assessed at about 96 weeks after the first infusion.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves a reduced total number of Gd-enhancing T1 lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • administration of the anti-CD20 antibody e.g., ublituximab
  • the method reduces the number of new or enlarging T2 hyperintense lesions in the subject following administration of the anti-CD20 antibody composition as described herein. In some embodiments, reduction of the number of new or enlarging T2 hyperintense lesions is assessed by MRI scan. In some embodiments, reduction of the number of new or enlarging T2 hyperintense lesions is assessed at about 96 weeks after the first infusion.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves a reduced total number of new and enlarging T2 hyperintense lesions per MRI scan, as compared to a subject that receive orally administered 14 mg of teriflunomide daily during the same treatment period.
  • administration of the anti-CD20 antibody e.g., ublituximab
  • a relative reduction of about 90-92% is a relative reduction of about 90-92%.
  • the method achieves confirmed disability progression in the subject following administration of the anti-CD20 antibody composition as described herein.
  • confirmation of disability progression comprises an increase of greater than or equal to 1.0 point from a baseline EDSS score of the subject attributable to MS, wherein said baseline EDSS score is 5.5 or less.
  • confirmation of disability progression comprises an increase of greater than or equal to 0.5 point from a baseline EDSS score of the subject attributable to MS, wherein said baseline EDSS score is greater than 5.5.
  • the method results in no evidence of disease activity (NED A) in the subject.
  • NED A comprises one or more of no confirmed relapses, no gadolinium-enhancing (Gd+) T1 lesions, no new and/or enlarging T2 lesions, and no 12-week confirmed disability progression.
  • methods of the present disclosure results in NED A in the subject about 24 weeks after administration of a pharmaceutical formulation (e.g., an anti-CD20 antibody composition) as described herein.
  • administration of a pharmaceutical formulation described herein can result in NEDA in the subject about 24-96 weeks (e.g., 24-48 weeks, 24-72 weeks, 48-72 weeks, 72-96 weeks, or 48-96 weeks (e.g., 24 weeks, 36 weeks, 48 weeks, 60 weeks, 72 weeks, 84 weeks, 96 weeks, or any range in between)) after the administration.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody e.g., ublituximab
  • administration of the anti-CD20 antibody increases NEDA status by 197%. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, increases NEDA status by 277%.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves increased Confirmed Disability Improvement (CDI), as compared to a subjects that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • CDI Confirmed Disability Improvement
  • administration of the anti-CD20 antibody e.g., ublituximab
  • administration of the anti-CD20 antibody e.g., ublituximab
  • administration of the anti-CD20 antibody (e.g., ublituximab) as compared to teriflunomide administration, increased CDI by 103% at 24 weeks.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves an increased Multiple Sclerosis Functional Composite (MSFC) score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • MSFC Multiple Sclerosis Functional Composite
  • administration of the anti-CD20 antibody e.g., ublituximab
  • teriflunomide administration increased the MSFC score by about 76%- 90%.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves an improved timed 25-Foot Walk (T25FW) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the subject administered the multiinfusion dosage regimen of the anti-CD20 antibody e.g., ublituximab
  • achieves an improved 9- Hole Peg test (9-HPT) Score as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves a significant reduction in both volume and number of new T1 hypointense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
  • the anti-CD20 antibody e.g., ublituximab
  • the treatment for MS further comprises administering the anti-CD20 antibody in combination with one or more additional therapeutic agents.
  • the anti-CD20 antibody used in the present methods of treating RMS can be given in combination with other compounds, drugs, and/or agents useful for the treatment of RMS.
  • Such compounds, drugs, and/or agents can include, for example, small molecule drugs, monoclonal antibodies, or other B-cell depleting agents.
  • the methods described herein are used in combination with current standard of care MS treatment.
  • the anti-CD20 antibody is used in combination with a Bruton’s tyrosine kinase (BTK) inhibitor for treating RMS, including, e.g., fenebrutinib, evobrutinib, tolebrutinib, orelabrutinib BIIB091, AC0058, PRN473 (Dolgin, Nature Biotechnology 39:3-12 (2021), as well as those compounds described in U.S. Patent No. 9,951,077.
  • the anti-CD20 antibody is used in combination regimens that take advantage of non-overlapping mechanisms from ublituximab for B-cell depletion.
  • the anti-CD20 antibody can be combined with injectable medications, oral medications, or infused medications.
  • injectable medications for combination therapy with the anti-CD20 antibody can include interferons, including but not limited to AVONEX (interferon beta-la), BETASERON (interferon beta-lb), EXTAYIA (interferon betalb), PLEGRIDY (peginterferon beta- la), and REBIF (interferon beta- la).
  • Other injectable medications can include COPAXONE (glatiramer acetate).
  • Orally available medications for combination with ublituximab can inlucde AUBAGIO (teriflunomide), fumarate based compositions including BAFIERTAM, dimethyl fumarate, TECFIDERA, and VUMERITY.
  • the anti-CD20 antibody e.g., ublituximab
  • the anti-CD20 antibody can be used in combination with the monoclonal antibody Tysabri (natalizumab).
  • Orally administered agents that can be used in combination with the anti-CD20 antibody include GILENYI (fingolimod), MAYZENT (Siponimod), ZEPOSIA (ozanimod), and PONVORY (ponesimod). Additional orally administered agents include MAVENCLAD (cladribine).
  • the anti-CD20 antibody e.g., ublituximab
  • infused medications including, LEMTRADE (alemtuzumab) and NOVANTRONE (mitoxantrone).
  • an anti-CD20 antibody of the present disclosure e.g., ublituximab or an anti-CD20 antiody that binds the same epitope as ublituximab
  • ublituximab targets a unique epitope region on the CD20 antigen that is not targeted by other anti-CD20 monoclonal antibodies.
  • additional therapeutic agents can comprises a B-cell depleting agent that is not an anti-CD20 antibody.
  • the B-cell depleting agent is a PI3K inhibitor.
  • the additional therapeutic agent comprises a B-cell depleting agent that, like the anti-CD20 antibody (e.g., ublituximab) described here, is an anti-CD20 antibody.
  • ublituximab can be used in combination with additional anti-CD20 antibody compositions, such as, e.g., OCREVUS® (ocrelizumab), KESIMPTA® (ofatumumab), and Rituxan (rituximab).
  • the additional therapeutic agent can be administered orally, parenterally, intravenously, or subcutaneously.
  • TEAE treatment-emergent adverse events
  • TEAE comprises cytopenia or reduction in blood cell count in the subject.
  • Blood cell count can be evaluated by blood tests, such as a complete blood count (CBC).
  • Blood cell count can be obtained by cell counting methods known in the art, including, but not limited to manual methods (e.g., by using a hemocytometer) and automated methods (e.g., by using an automated cell counter).
  • cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60%, about 50-70%, about 60-70%, about 60-80%, about 70-80%, about 70-90%, about 80-90%, about 80-100%, about 90-100%, or any range in between (e.g., about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, about 100%, or any range in between)) reduction or decrease in blood cell count in a subject, as compared to a control or normal blood cell count.
  • cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60
  • a control or normal blood cell count may comprise the blood cell count of a healthy subject who has not been administered the pharmaceutical formulation of the present disclosure. Additionally, or in the alternative, blood cell count of a subject prior to treatment by the present methods may serve as the control or normal blood cell count. Cytopenia or reduction in blood cell count can comprise one or more of lymphopenia, neutropenia, leukopenia, and anemia.
  • administration of a pharmaceutical formulation results in cytopenia in the subject about 1-3 days (e.g., about 1 day, about 2 days, or about 3 days) after the administration.
  • administration of the pharmaceutical formulation can result in lymphopenia in the subject about 2 days after the administration.
  • the cytopenia e.g., lymphopenia, neutropenia, leukopenia, and/or anemia
  • the cytopenia is transient, such that blood cell count in the subject is normalized (e.g., becomes same or similar to control or normal blood cell count) by about 15 days (e.g., by 14 days, by 13 days, by 12 days, by 11 days, by 10 days, by 9 days, by 8 days, by 7 days, by 6 days, by 5 days, or by 4 days) after administration of the pharmaceutical formulation.
  • lymphocyte count in the subject can be normalized in the subject by 8 days after administration of the pharmaceutical formulation.
  • the cancer is a bladder cancer, a breast cancer, a cervical cancer, a colorectal cancer, a gynecologic cancer (i.e., cervical, ovarian, uterine, vaginal, or vulvar cancer), head and neck cancer, kidney cancer, liver cancer, lung cancer, lymphoma, mesothelioma, myeloma, prostate cancer, skin cancer, or thyroid cancer.
  • a gynecologic cancer i.e., cervical, ovarian, uterine, vaginal, or vulvar cancer
  • head and neck cancer kidney cancer, liver cancer, lung cancer, lymphoma, mesothelioma, myeloma, prostate cancer, skin cancer, or thyroid cancer.
  • the disease or disorder associated with excessive B-cell proliferation is a hematological cancer.
  • the hematological cancer is lymphoma, leukemia, or myeloma.
  • the hematological cancer is selected from B-cell lymphoma, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma (FL), Waldenstrom's macroglobulinemia (WM), diffuse large B-cell lymphoma (DLBCL), marginal zone lymphoma (MZL), hairy cell leukemia (HCL), Burkitt's lymphoma (BL), Richter's transformation, or primary central nervous system lymphoma (PCNSL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MM multiple myelo
  • the anti- CD20 antibody composition as described herein comprises anti-CD20 antibodies, which are IgGl antibodies comprising heavy chains each comprising the amino acid sequence of SEQ ID NO:1, and light chains each comprising the amino acid sequence of SEQ ID NO:2.
  • the hematological cancer is B-cell lymphoma. In some embodiments, the B-cell lymphoma is relapsed or refractory. In certain embodiments, the hematological cancer is non-Hodgkin's lymphoma (NHL). In some embodiments, the NHL is relapsed or refractory. In certain embodiments, the hematological cancer is Waldenstrom's macroglobulinemia (WM). In some embodiments, the WM is relapsed or refractory. In certain embodiments, the hematological cancer is marginal zone lymphoma (MZL). In some embodiments, the MZL is relapsed or refractory.
  • the hematological cancer is chronic lymphocytic leukemia (CLL). In some embodiments, the CLL is relapsed or refractory. In certain embodiments, the hematological cancer is small lymphocytic lymphoma (SLL). In some embodiments, the SLL is relapsed or refractory. In certain embodiments, the hematological cancer is primary central nervous system lymphoma (PCNSL). In some embodiments, the PCNSL is relapsed or refractory.
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • PCNSL primary central nervous system lymphoma
  • the method of treating a hematological cancer comprises administering to the subject the anti-CD20 antibody composition as described herein by intravenous infusion.
  • the intravenous infusion comprises: a) an infusion comprising about 900 mg of the anti-CD20 antibody composition as described herein on days 1 and 2 (split into a 150 mg dose on day 1 and a 750 mg dose on day 2), day 8, and day 15 of Cycle 1 (each Cycle being 28 days); day 1 of Cycles 2-6; and day 1 of every 3 cycles after Cycle 6 (e.g., Cycle 9, 12, 15, etc.).
  • the hematological cancer is CLL.
  • TEAE treatment-emergent adverse events
  • TEAE comprises cytopenia or reduction in blood cell count in the subject.
  • Blood cell count can be evaluated by blood tests, such as a complete blood count (CBC).
  • Blood cell count can be obtained by cell counting methods known in the art, including, but not limited to manual methods (e.g., by using a hemocytometer) and automated methods (e.g., by using an automated cell counter).
  • cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60%, about 50-70%, about 60-70%, about 60-80%, about 70-80%, about 70-90%, about 80-90%, about 80-100%, about 90-100%, or any range in between (e.g., about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, about 100%, or any range in between)) reduction or decrease in blood cell count in a subject, as compared to a control or normal blood cell count.
  • cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60
  • a control or normal blood cell count may comprise the blood cell count of a healthy subject who has not been administered the pharmaceutical formulation of the present disclosure. Additionally, or in the alternative, blood cell count of a subject prior to treatment by the present methods may serve as the control or normal blood cell count. Cytopenia or reduction in blood cell count can comprise one or more of lymphopenia, neutropenia, leukopenia, and anemia.
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1; b) a second infusion comprising about 500 to about 700 mg, about 800 to about 1000 mg, or about 1100 to about 1300 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 500 to about 700 mg, about 800 to about 1000
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosy lated glycans) at day 1 ; b) a second infusion comprising 600, 900, or 1200 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising 600, 900, or 1200 mg of the anti- CD20 antibody composition at about 2 weeks after the first infusion; d) a fourth infusion comprising 600, 900,
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 500 to about 700 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 800 to about 1000 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent in
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising 600 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising 900 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusions comprising 450, 600, or
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 5 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; and b) one or more subsequent infusions comprising about 5 to about 450 mg of the anti-CD20 antibody composition at about one week after the prior infusion, wherein each subsequent infusion is at higher dose than the prior infusion.
  • the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; and b) one or more subsequent infusions comprising about 400 to about 500 mg of the anti-CD20 antibody composition at about one week after the prior infusion.
  • the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over 1 hour.
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody composition at day 1; and b) one or more subsequent infusions comprising 450 mg of the anti- CD20 antibody composition at about one week after the prior infusion.
  • the method comprises seven or more subsequent infusions.
  • the method comprises seven subsequent infusions.
  • the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over 1 hour.
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody at about 1 week after the first infusion; c) a third infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusion
  • the one or more subsequent infusions each comprise 600 mg of the anti-CD20 antibody composition as described herein.
  • the first infusion, second infusion, third infusion, and one or more subsequent infusions each comprise 600 mg of the anti-CD20 antibody composition as described herein.
  • the first infusion, second infusion, third infusion, and one or more subsequent infusions each comprise 900 mg of the anti-CD20 antibody composition as described herein.
  • the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
  • the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 700 to about 800 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 850 to about 950 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more
  • the first infusion comprises 150 mg of the anti-CD20 antibody composition
  • the second infusion comprises 750 mg of the anti-CD20 antibody composition
  • the third and one or more subsequent infusions comprise 900 mg of the anti-CD20 antibody composition.
  • the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
  • the subject is a human. In some embodiments, the human subject is an adult. In some embodiments, the subject has relapsed or refractory B-cell lymphoma. In some embodiments, the subject has been previously treated with at least one prior course of rituximab or a rituximab-based therapy. In some embodiments, the subject has been previously treated with at least one prior course of fludarabine or a fludarabine-based therapy. In some embodiments, the subject is naive to treatment for a B-cell lymphoma. In some embodiments, the subject is eligible for high dose or combination chemotherapy and/or stem cell transplant.
  • the subject has an Eastern Cooperative Oncology Group (ECOG) score of from 0 to 2 prior to treatment with the anti-CD20 antibody composition as described herein. In some embodiments, the subject has a peripheral blood lymphocyte count of greater than 5,000/pL prior to treatment with the anti-CD20 antibody composition as described herein.
  • ECOG Eastern Cooperative Oncology Group
  • the anti-CD20 antibody compositions provided herein can be used to treat and/or prevent Chronic inflammatory demyelinating polyneuropathy (CIDP); Myositis; Lupus Nephritis; other forms of MS- PPMS, SPMS; Myasthenia Gravis (MG); Antiphospholipid Syndrome; Thrombotic Thrombocytopenic Purpura (TTP); Ulcerative Colitis; Minimal Change Nephrotic Syndrome (MCNS); Aplastic Anemia; Autoimmune Glomerulopathies; Rheumatoid Arthritis (RA); Interstitial Lung Disease; Myasthenia Gravis (MG); Subepidermal Autoimmune Blistering Disease; Pulmonary Infectious Diseases; Acquired Hemophilia; Refractory Mixed Cryoglobulinemia; Primary Immune Thrombocytopenia; Graft Versus Host Disease (GVHD); Autoimmune Blistering Disease; Anti-Myelin Associated Glyco
  • CIDP Chronic inflammatory dem
  • the anti-CD20 antibody are administered as i) a first infusion at a dose of about 150 mg, ii) a second infusion two week later at a dose of about 450 mg, and iii) subsequent infusions every six months at a dose of about 450 mg.
  • the administration of the anti-CD20 antibody produces an area under the curve (AUC) of between about 2,160 pg/mL and about 3,840 pg/mL. In certain embodiments, the AUC is about 3,000 pg/mL. In certain embodiments, the AUC is the steady state AUC.
  • the administration of the anti-CD20 antibody produces a Cmax of between about 118,011 ng/mL and about 159,989 ng/mL. In certain embodiments, the Cmax is about 139,000 ng/mL. In certain embodiments, the Cmax is the steady state Cmax.
  • the administration of the anti-CD20 antibody produces a Cmin of about 0 ng/mL and about 375 ng/mL. In certain embodiments, the Cmin is about 139 ng/mL. In certain embodiments, the Cmin is the steady state Cmin.
  • the administration of the anti-CD20 antibody produces a Cavg of between about 6,437 ng/mL and about 11,443 ng/mL. In certain embodiments, the Cavg is about 8,940 ng/mL. In certain embodiments, the Cavg is the steady state Cavg.
  • the autoimmune disease is selected from the group consisting of multiple sclerosis, psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, osteoporosis, eczema, allogenic or xenogenic transplantation (organ, bone marrow, stem cells and other cells and tissues), graft rejection, graft-versus-host disease, lupus erythematosus, inflammatory disease, type 1 diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis (e.g., Hasimoto's and autoimmune thyroiditis), myasthenia gravis, autoimmune hemolytic anemia, cystic fibrosis, chronic relapsing hepatitis, primary biliary cir
  • the autoimmune disease is multiple sclerosis.
  • the multiple sclerosis is a relapsing form of multiple sclerosis.
  • the relapsing form of multiple sclerosis is a clinically isolated syndrome (CIS); relapsing-remitting MS (RRMS); active secondary progressive MS (SPMS); or primary progressive MS (PPMS).
  • the relapsing form of MS is a clinically isolated syndrome (CIS).
  • the relapsing form of MS is relapse-remitting multiple sclerosis (RRMS).
  • the relapsing form of MS is active secondary progressive multiple sclerosis (SPMS).
  • the relapsing form of MS is primary progressive MS (PPMS).
  • the anti-CD20 antibody is administered intravenously.
  • Example 15 provides exemplary methods of making the populations of anti-CD20 antibody proteins provided herein.
  • the methods of making the populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications as described above comprises: i) culturing the rat hybridoma cells at a first culture pH of about 7.0 to about 7.55 for 0 to 3 days, ii) culturing the rat hybridoma cells at a second culture pH of about 6.5 to about 6.99 on day 3, iii) maintaining the culture pH at the second culture pH of about 6.5 to about 6.99 from culture day 3 until day 14 of the cell culture, and iv) controlling culture pCCh levels to less than about 200 mmHg throughout the culture period.
  • the second culture pH is about 6.60 to about 6.96 (e.g., the second culture pH is 6.8).
  • the second culture pH results in higher integrated viable cell density (IVCD) and higher titer at harvest.
  • IVCD integrated viable cell density
  • the second culture pH results in lower percent fucosylation.
  • the rat hybridoma cells expressing the recombinant protein are cultured in a basal media that is chemically defined and animal-derived component free (ADCF) culture medium
  • the basal medium is supplemented with a feed medium.
  • the method further comprises an initial temperature set point of about 37°C, wherein said initial temperature set point is set on culture day 0 to culture day 1.
  • the method further comprises a second temperature set point of about 35°C, wherein said second temperature set point is set at the end of culture day 1 to culture day 3.
  • the method further comprises comprising a third temperature set point of about 32°C to about 33°C, wherein said third temperature set point is set on culture day 3 and is maintained though harvest.
  • harvest refers to the point of time in the mammalian cell culture process when cells containing the recombinant protein are separated and removed from the cell culture media and subject to additional processing, such as, e.g., centrifugation, filtration, or purification.
  • harvest of the cells will occur on process day 12, 13, or 14 of the cell culture, or when cell viability drops below 20%, whichever comes first.
  • the method further comprises harvesting the recombinant protein produced by the rat hybridoma cell.
  • the method further comprises purifying the recombinant protein by affinity chromatography and/or ion exchange chromatography.
  • the affinity chromatography comprises protein A purification.
  • the methods result in an increased yield of recombinant protein.
  • the recombinant protein is increased by at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 110 %, at least about 120%, at least about 130%, at least about 140%, or at least about 150%, relative to a recombinant protein produced by a culturing process that does not employ the culture conditions as recited above.
  • the ublituximab infusions are prepared in 250 mb of 0.9% Sodium Chloride Injection.
  • ublituximab infusions can be prepared in infusion bags containing 0.9% Sodium Chloride Injection.
  • the first infusion of 150 mg ublituximab is prepared by: (1) inspecting one vial of 150 mg/6 mL of ublituximab solution for any particulate matter or discoloration, and not using the solution if the solution contains discrete foreign particulate matter; (2) if no particulate matter in said vial, preparing 250 mL infusion bag for first infusion by using one vial of 150 mg/6 mL ublituximab solution; (3) withdrawing 6 mb 0.9% Sodium Injection from the infusion bag, and discarding; (4) withdrawing 6 mL ublituximab from said vial; (5) diluting 6 mL (150 mg) of ublituximab into said 250 mL infusion bag containing 0.9% Sodium Injection for immediate administration; and (6) mixing said infusion bag by gentle inversion, without shaking.
  • the second or subsequent infusions of 450 mg ublituximab are prepared by: (1) inspecting three vials of 150 mg/6 mL of ublituximab solution for any particulate matter or discoloration, and not using the solution if the solution contains discrete foreign particulate matter; (2) if no particulate matter in said vial, preparing 250 mL infusion bag for second or subsequent infusion by using three vials of 150 mg/6 mL ublituximab solution; (3) withdrawing 18 mL 0.9% Sodium Injection from the infusion bag, and discarding; (4) withdrawing 18 mL ublituximab from said vial; (5) diluting 18 mL (450 mg) of ublituximab into said 250 mL infusion bag containing 0.9% Sodium Injection for immediate administration; and (6) mixing said infusion bag by gentle inversion, without shaking.
  • the content of the infusion bag is at room temperature.
  • the subject prior to intitiating ublituximab administration, the subject is pre-screened for Hepatitis B virus (HBV). If the subject tests positive for Hepatitis B surface antigen (HbsAg), ublituximab should not be administered. In some embodiments, the subject being administered the ublituximab infusion is hepatitis B virus surface antigen (HBsAg) negative.
  • HBV Hepatitis B virus
  • HbsAg Hepatitis B virus surface antigen
  • the subject is pre-medicated with a corticosteroid and an antihistamine, 30-60 minutes prior to administration of ublituximab or an anti-CD20 antibody that binds the same epitope as ublituximab, both of which may be administered orally or intravenously.
  • the corticosteroid and/or antihistamine are administered to the subject orally.
  • the pre-treatment dosage of corticosteroid is about 100 mg methylprednisone, 10-20 mg dexamethasone, or an equivalent corticosteroid.
  • ublituximab is administered in a multi-dosage regimen.
  • ublituximab is administered by intravenous infusion.
  • ublituximab can be administered in a multi-infusion dosage regimen by intravenous infusion.
  • ublituximab is administered to a patient with RMS by intravenous infusion in a multi-infusion dosage regimen for 48 weeks.
  • ublituximab is administered to a patient with RMS by intravenous infusion in a multi-infusion dosage regimen for 96 weeks.
  • the multi-infusion dosage regimen can comprise first, second, and subsequent intravenous infusions of ublituximab.
  • “subsequent infusions” of ublituximab can be any number of infusions after the first and second infusion.
  • the multi-infusion dosage regimen comprises first and second infusions of 150 mg ublituximab intravenous infusion (First Infusion), followed two weeks later by a 450 mg ublituximab intravenous infusion (Second Infusion). In some embodiments, the multi-infusion dosage regimen further comprises subsequent infusions of 450 mg ublituximab intravenous infusion every 6 months.
  • the duration of the first infusion of ublituximab, or an anti- CD20 antibody that binds the same epitope as ublituximab is about four hours.
  • the infusion rate of the first infusion of ublituximab, or an anti-CD20 antibody that binds the same epitope as ublituximab can be 10 mb per hour for the first 30 minutes; 20 mb per hour for the next 30 minutes; 35 mb per hour for the next hour; and 100 mb per hour for the remaining two hours.
  • the duration of the second and subsequent infusions of ublituximab is about one hour.
  • the infusion rate of the second and/or subsequent infusion of ublituximab can be 100 mb per hour for the first 30 minutes, and 400 mb for the remaining 30 minutes.
  • the infusion duration may take longer if infusion is interrupted or slowed.
  • the subject is monitored for at least one hour after the completion of the first two ublituximab infusions. Subsequent infusions do not require monitoring post-infusion unless an infusion-related reaction (IRR) and/or hypersensitivity has been observed.
  • IRR infusion-related reaction
  • the ublituximab intravenous infusion is stopped and permanently discontinued if the subject has symptoms of a life-threatening infusion-related reaction. In some embodiments, the ublituximab intravenous infusion is discontinued if the subject has symptoms of a severe infusion-related reaction, and restarted once the subject’s infusion-related reaction symptoms have resolved. In some embodiments, the ublituximab intravenous infusion is restarted at half the infusion rate at the time of onset of the infusion- related reaction. The ublituximab infusion rate can be increased to the original infusion rate if the subject tolerates the half infusion rate.
  • the ublituximab infusion rate is reduced in half if the subject has symptoms of a mild to moderate infusion-related reaction, wherein the half infusion rate is maintained for at least 30 minutes.
  • the ublituximab infusion rate can be increased to the original infusion rate if the subject tolerates the half infusion rate.
  • the administration of ublituximab can be via routes of admistration other that intravenous infusion (e.g., subcutaneous injection, intramuscular injection, oral, epidermal, spinal, or inhalation).
  • intravenous infusion e.g., subcutaneous injection, intramuscular injection, oral, epidermal, spinal, or inhalation.
  • composition can refer to a composition that is acceptable for pharmaceutical administration, such as to a human being.
  • a composition can include substances that are impurities at a level not exceeding an acceptable level for pharmaceutical administration (such level including an absence of such impurities), and can include pharmaceutically acceptable excipients, vehicles, carriers, stabilizers, and other inactive ingredients, for example, to formulate such composition for ease of administration, in addition to any active agent(s).
  • compositions comprising ublituximab, or an antibody that binds the same epitope as ublituximab, for use in the treatment of subjects with relapsing forms of Multiple Sclerosis, according to any of the methods disclosed herein.
  • the pharmaceutical composition comprises ublituximab.
  • the pharmaceutical composition comprising ublituximab is formulated together with a pharmaceutical carrier. Suitable pharmaceutical carriers are known to those skilled in the art (Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA (1990)).
  • the pharmaceutical composition can comprise any number of excipients.
  • Excipients that can be used include carriers, surface active agents, thickening or emulsifying agents, solid binders, dispersion or suspension aids, solubilizers, colorants, flavoring agents, coatings, disintegrating agents, lubricants, sweeteners, preservatives, isotonic agents, and combinations thereof.
  • the selection and use of suitable excipients is taught in Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Ed. (Lippincott Williams & Wilkins 2003), the disclosure of which is incorporated herein by reference.
  • compositions described here can be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion).
  • the active compound can be coated in a material to protect it from the action of acids and other natural conditions that may inactivate it.
  • parenteral administration can mean modes of administration other than enteral and topical administration, usually by injection, and can include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • compositions described here can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
  • a non-parenteral route such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
  • the pharmaceutical composition comprising ublituximab is for administration to a subject by intravenous infusion.
  • the glycosylation profile of a sample of anti-CD20 antibody proteins provided herein was determined by measuring fluorescently labeled N-glycans (fluorescent label is 2- aminobenzamide) that were enzymatically cleaved from the anti-CD20 antibody proteins using PNGase F.
  • the labeled glycans are resolved using a hydrophilic interaction column equipped.
  • the glycans flow through a fluorescence detector after separation. Peak identification from the test sample chromatograms is identified based on retention time and relative to peaks in the glycan standard which have been confirmed by mass spectrometry.
  • the relative percentage of each N-glycan is calculated based on the N-glycan peak area divided by the total peak area of all N-glycans.
  • the glycosylation profile is shown in Figure 2.
  • the glycosylation profile of anti-CD20 antibody proteins provided herein was assessed by intact mass analysis (LC-MS) under non-reducing conditions.
  • the sample of anti- CD20 antibody proteins provided herein is first exchanged into MS appropriate buffers during the chromatography step using SEC and a mobile phase containing TFA, acetonitrile, and water.
  • the sample is then introduced into an ESI-QTOF for intact mass analysis.
  • the mass spectra are deconvoluted and the peaks are assigned based on mass.
  • the relative abundance of each anti- CD20 antibody proteins provided herein containing N-glycan was calculated by taking the abundance of an N-glycan and dividing by the total abundance of all identified peaks. Results are provided in the table below and in Figure 3.
  • Antibody dependent cytotoxicity is mediated through binding of the Fc portion of TG-1101 (TG Therapeutics, Inc.) to the FcylllA Receptor on the effector cells.
  • the assay used for this analysis employs Eurofins- DiscoverX’s “KILR CD 16a effector cells”, which are single donor-derived human CD8+ T-lymphocytes engineered to express CD16 (FcyRIII) on their plasma membrane surface.
  • KILR CD 16a effector cells are single donor-derived human CD8+ T-lymphocytes engineered to express CD16 (FcyRIII) on their plasma membrane surface.
  • cytotoxic T cells provide reduced background killing, increased accuracy and reproducibility compared to PMBC preparations isolated from fresh blood.
  • Raji cells are used as target cells, and ADCC activity is determined from the lysis of the target cells.
  • KILR cells are obtained from Eurofins, and Raji cells from ATCC. Master and Working cell bank system was used to ensure quality of the reagents. Raji cells were seeded at 1x10 5 cells/mL, KILR Effector cells were seeded at 5x10 5 cells/mL, and final effector: target (E:T) ratio was 5:1. An eight-point dilution series of the samples were used in the concentration range of 250.00 pg/ml - 0.04 pg/ml (250, 50, 16.7, 5.6, 1.9, 0.6, 0.2, 0.04 pg/ml). Cell mixtures and testing samples were cultured at 36 ⁇ 1°C, 5 ⁇ 1%CO2 for 18-22 hours.
  • a CytoTox GLoTM preparation is added, and plates are incubated for 30 ⁇ 10 minutes. The plates are read using the SpectraMax plate reader. Two independent preparations of the materials are prepared and assayed across duplicate plates. Assay controls are prepared in triplicates and include: Target Cells Alone Control, Target Cells Death Control, Effector Cells Alone Control, and Effector & Target Cells Control. SoftMax Pro was used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. Results are reported as percentage ADCC activity compared to Ublituximan reference standard. EC50 of each testing sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP482, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra.
  • TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 4, all anti-CD20 samples exhibited dose- dependent ADCC activity, TG-1101 (TG Therapeutics, Inc.) has highest ADCC activity compared to other anti-CD20.
  • the ADCC activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 9, compared to TG-1101 (TG Therapeutics, Inc.), bedroomsva has ADCC activity that is relative similar to TG-1101 (TG Therapeutics, Inc.), while the ADCC activities of Rituxan, Ocrevus, and Arzerra are significantly lower.
  • the ED50s of each anti-CD20 are also shown in Table 9, overall TG-1101 (TG Therapeutics, Inc.) and Gazyva have lower EC50 than Arzerra, Rituxan, and Ocrevus. EC50 of TG-1101 (TG Therapeutics, Inc.) is approximately 25 fold lower than Ocrevus.
  • This ADCC assay was performed using CD20 expressing Raji cells as target cells, primary NK cells as effector cells, and LDH as target cell lysis read-out.
  • Raji cells (ATCC, Cat# CCL-86TM) were seeded atlxl05 cells/well on plates.
  • Primary NK cells isolated from human donor PBMC using Miltenyibiotec kit (Cat# 130-092-657) . E/T ratio of 5: 1 for NK92/CD16a cells and primary NK cells was used in the assay.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, and Arzerra. As shown in Figure 5, all anti-CD20 samples exhibited dose-dependent ADCC activity. The calculated EC50 values are summarized in Table 10. TG-1101 (TG Therapeutics, Inc.) and Gazyva displayed higher ADCC activities and lower EC50 than Rituxan and Arzerra.
  • ADCP Antibody dependent cellular phagocytosis
  • MO As Antibody dependent cellular phagocytosis
  • ADCP activity was assessed using an assay in which CD20 expressing Daudi cells were used as target cells (ATCC, Cat# CCL-213, labeled by PKH26).
  • Human monocytes were isolated from PBMC from 20 human donors (using human Pan Monocyte Isolation Kit, MiltenyiB iotec, Cat#130-096-537) and differentiated in vitro using GM- CSF to yield macrophages. An E/T ratio of 5:1 was used; and the ADCP was assessed by flow cytometry in this assay.
  • ADCP Effector and target cells control with a non-specific IgGl antibody; Effector and target cells control (background control).
  • ADCP was determined by FACS as a percentage of PKH26/PKH67 double positive cell counts/PKH26.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, and Arzerra. As shown in Figure 6, all anti-CD20 samples exhibited dose-dependent ADCP activity. The calculated EC50 values are summarized in Table 11. The EC50s are in the ng/ml range, and given the nature of the test method, the ADCP activities of all samples are considered similar.
  • the Complement Dependent Cytotoxicity is mediated through binding of the Fc portion of TG-1101 (TG Therapeutics, Inc.) to Cl q Receptor in the complement system.
  • the CDC activity assay used in this analysis is a cell-based assay using the CD20 expressing human mantle cell lymphoma cell line, Jeko-1 and rabbit serum as the source of complement. CDC mediated cell lysis is measured by the Cell Titer-GioTM reagent (Promega).
  • a nine-point dilution series of the samples are used in the concentration range of 10,000 ng/ml - 10.42 ng/ml (10,000.00, 3333.33, 1666.67, 833.33, 416.67, 208.33, 104.17, 52.08, 10.42 ng/ml).
  • Two independent preparations of each sample are prepared and assayed across duplicate plates.
  • Assay negative controls are prepared in triplicates and include target cells & complement control and target cells alone control.
  • Jeko-1 cells obtained from the ATCC and maintained through a master banking system, were seeded at 3x10 5 cells/mL and incubated for 60-90 minutes. Samples dilutions, and then complement were added, and the plates were incubated for approximately 2 hours at 37°C and 25 minutes at room temperature. Target cells with complement only control and target cell only control provided a basal level of target cell viability over the course of the assay. The Cell Titer-Gio reagent is then added and incubated an additional 30 minutes at room temperature. At the end of the assay the plates are read using a SpectraMax M5 plate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit.
  • TG-1101 TG Therapeutics, Inc.
  • Gazyva TG-1101
  • Rituxan Ocrevus
  • Arzerra TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 7, all samples exhibited dose-dependent CDC activity, except for Gazyva, which is known to have reduced CDC activity.
  • Rituxan and Arzerra have comparable CDC activity; Ubli and Ocrevus have comparable CDC activity.
  • the CDC activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 12, compared to TG-1101 (TG Therapeutics, Inc.), lendersva has ADCC activity that is relative similar to TG-1101 (TG Therapeutics, Inc.), while the ADCC activities of Rituxan, Ocrevus, and Arzerra are significantly lower.
  • the ED50s of each anti-CD20 are also shown in Table 12, which shows similar comparisons as the level of CDC activities.
  • the CD20 binding used for this analysis is a cell based binding assay that uses a CD20 expressing human mantle cell lymphoma cell line, Jeko-1, and an MSD (MesoScale Discovery) assay format. Jeko-1 target cells are seeded onto MSD plates, test samples are incubated and allowed to bind to Jeko-1 cells, anti- human Fc detection antibody conjugated with streptavidin-SULFOTAGTM is used to emit electrochemiluminescence signal.
  • MSD MesoScale Discovery
  • test samples were used in the concentration range of 40,000.00 pg/ml - 0.23 ng/ml (40,000.00, 4,000.00, 1,000.00, 333.30, 111.10, 37.00, 4.60, 0.23 ng/ml).
  • Assay controls include No cell control (Reference Standard/Test Material dilution + detection reagent, omitting cells) and Cell only control (Cells + detection reagent, omitting Reference Standard/Test Material).
  • Jeko-1 cells obtained from the ATCC and managed through a master banking system, are seeded onto MSD high bind plate in PBS at 3 x 10 5 cells per mL, in a final volume of 100 pL per well, and incubated at 35-37°C for 2 hours ⁇ 10 min. Unbound cells are removed by a PBS wash, the plates are blocked then washed. Fifty pL of sample dilutions were added, and the plate is incubated at room temperature for 1 hour ⁇ 10 min while shaking. Following incubation and three washes, 50 pL anti-human Fc detection antibody conjugated with STREP-SULFOTAG is added and incubated for 1 hour ⁇ 10 min at room temperature while shaking.
  • the plates are washed again, and 150 pL of the MesoScale read buffer, containing tripropylamine (TP A), is added as a co-reactant for light generation for an electrochemiluminescence read out. Plates are read immediately on a MSD Reader using Workbench 4.0. The resulting data is evaluated using the PLA software and analyzed using a constrained 4 parameter logistic model. Binding activity results are reported as percentage potency relative to the reference standard. EC50 of each testing sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP466, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
  • TP A tripropylamine
  • TG-1101 TG Therapeutics, Inc.
  • Gazyva which is a Type II anti-CD20 and is known to have approximately 50% of target occupancy
  • Ubituximab, Rituxan, Ocrevus, and Arzerra have similar maximum binding.
  • the CD20 binding activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard and binding EC50s are shown in Table 13.
  • the CD20 binding affinity of the 4 anti-CD20s are similar. Table 13: CD20 Binding Activity and EC50
  • EC50s values listed are negative, rising from the PLA software log transforms the concentrations to base 2.
  • TG-1101 TG Therapeutics, Inc.
  • TG Therapeutics, Inc. The binding of TG-1101 (TG Therapeutics, Inc.) to cell surface CD20 on Raji and Daudi cells was characterized by FACS analysis at LakePharma. A six-point dilution series of the samples, in duplicates and in the concentration range of 40 pg/ml - 0 ug/ml with a dilution factor of 5 were used. Cells were incubated with sample dilutions; binding was detected using a PE conjugated anti-human secondary antibody.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Arzerra, and Rituxan. As shown in Figure 9, all anti-CD20 samples exhibited dose-dependent CD20 binding to both Daudi and Raji cells. The binding characteristics as evaluated by the FACS assay are similar to that evaluated by the MSD assay. With the exception of Gazyva, which is a Type II anti-CD20 and is known to have approximately 50% of target occupancy, Ubituximab, Rituxan, and Arzerra have similar maximum binding. The calculated EC50 values are summarized in Table 14. The CD20 binding affinity of the four anti-CD20 antibodies are similar. Table 14: EC50 of Cell Surface CD20 Binding by FACS
  • the assay used in this analysis is a surface plasmon resonance (SPR)-based method that measures the binding to both FcyRIIIa 158V and FcyRIIIa 158F receptors.
  • SPR surface plasmon resonance
  • the method follows a direct binding assay methodology where the FcyRIIIa receptor is directly immobilized onto the flow cell on a sensor chip surface and samples are injected over the chip to assess binding.
  • FcyRIIIa 158V receptor (3pg/ml) or FcyRIIIa 158F receptor (6 pg/ml) is immobilized on the chip surface using covalent amine coupling chemistry.
  • Eight-point dilution series of the testing samples are prepared in the concentration range of 1000 nM - 15.6 nM with a dilution factor of 2. Independent duplicates of sample dilutions are injected over the chip, followed by surface regeneration between each cycle. The binding is measured in response units (RU). The kinetics of the binding reaction is determined by measuring changes in SPR due to the increase in mass in the close proximity to the biosensor chip surface. Change in the mass of the complex as a function of time is visualized as a sensorgram.
  • the equilibrium dissociation constants (KD) of each sample is determined for each receptor.
  • the rates of change of the SPR signal is analyzed using a 1 : 1 Langmuir model for FcyRIIIa 158V variant to yield apparent rate constants for the association and dissociation phases of the reaction, and equilibrium dissociation constants.
  • KD is determined using steady state affinity for the FcyRIIIa 158F variant.
  • the binding signals are exported into PLA to determine the relative binding response, elative affinity and relative binding for samples are also reported relative to the TG-1101 (TG Therapeutics, Inc.) reference standard. This test method is a validated assay, CTSOP477, used for TG-1101 (TG Therapeutics, Inc.) drug substance release testing. (b) Results
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Table 15, among the anti-CD20 antibodies tested, TG-1101 (TG Therapeutics, Inc.) has the highest binding affinities to both FcyRIIIa 158V and FcyRIIIa 158F receptors. Gazyva ranks the 2nd in binding affinities. For the high affinity receptor FcyRIIIal58V, TG-1101 (TG Therapeutics, Inc.) has ⁇ 15 folder higher affinity than Ocrevus; for the low affinity receptor FcyRIIIal58F TG-1101 (TG Therapeutics, Inc.) has ⁇ 10 folder higher affinity than Ocrevus.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Table 17, among the anti-CD20 antibodies tested, TG-1101 (TG Therapeutics, Inc.) has the highest binding affinities to both FcyRIIIa 158V and FcyRIIIa 158F receptors, corroborating the SPR data. Binding affinities to FCRN, which can affect PK, are similar among all the anti-CD20s. Compare to Ocrevus, TG-1101 (TG Therapeutics, Inc.) also has higher affinities for FcyRIIA and FcyRIIIB.
  • the Clq binding assay used for this analysis is an ELISA assay. Samples were coated on ELISA plates, HRP conjugated human Clq is incubated with samples on the plate. The bound HRP, in the presence of substrate TMB, generates a colorimetric signal. A 7-point dilution series of Test Materials were prepared in the concentration range of 15.00 ug/ml - 0.12 ug/ml with a dilution factor of 2. Sample dilutions are coated onto ELISA plates, and the plates incubated for 1 hour ⁇ 30 min at room temperature. After coating, the plates are washed, blocked, and washed again.
  • Peroxidase conjugated Clq was added, and the plates incubated for 1.5 hours ⁇ 30 minutes at room temperature. After incubation and washing, a tetramethylbenzidine (TMB) substrate solution was added and plates incubated at room temperature for 7 minutes (-1 min/ ⁇ 30 seconds). This produces a colorimetric reaction which is proportional to the level of Clq bound. The reaction is stopped with the addition of IM sulfuric acid and the color is measured at 450 nm using a Molecular Devices SpectraMax microplate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. Binding activity results are reported as percentage potency relative to the TG-1101 (TG Therapeutics, Inc.) reference standard.
  • EC50 of each test sample were generated for additional information.
  • This test method is a validated assay, CTSOP455, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
  • TG-1101 TG Therapeutics, Inc.
  • Gazyva TG Therapeutics, Inc.
  • Rituxan Ocrevus
  • Arzerra TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 10, Gazyva has minimal Clq binding (as expected), and the other 4 anti-CD20s exhibit dose dependent Clq binding.
  • TG-1101 (TG Therapeutics, Inc.) has slightly higher maximum binding.
  • the Clq binding activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 18.
  • TG-1101 (TG Therapeutics, Inc.) was characterized in an autologous normal human whole blood B-cell depletion assay in comparison to Gazyva, Rituxan, Ocrevus, and Arzerra.
  • Whole blood from three human donors was used, and donors were selected to have the 158V/158V genotype for FCGR3A_SNP target (rs396991).
  • B- cell depletion was evaluated by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3-positive T cells, CD19-positive B-cells, and CD20-positive B-cells. Percent of B-cell depletion (100-([100/B-/T-cell ratio in sample without antibody] x [B-/T-cell ratio in sample containing antibody])) was calculated and plotted against sample concentration.
  • Samples tested include TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808), A drug substance batch manufactured by the commercial process (C2) at Samsung Biologies (PPQ1), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Figures 11A-C, all samples exhibited dose-dependent B-cell depletion activity, even though there are some minor donor to donor differences. Overall, TG-1101 (TG Therapeutics, Inc.) and Gazyva have higher B cell depletion activities than Arzerra, Rituxan, and Ocrevus. Three anti- CD20 antibodies and one anti-CD19 antibody were used in this experiment.
  • TG-1101 TG Therapeutics, Inc.
  • Gazyva also have lower EC50 than Arzerra, Rituxan, and Ocrevus.
  • EC50 of TG-1101 is more than tenfold lower than Ocrevus.
  • the Cmax was obtained by the measurement of the highest point during the time of observation of drug concentration following the end of the absorption phase and the beginning of the elimination phase.
  • the Cmin was obtained by the measurement of the highest point during the time of observation of drug concentration following the end of the absorption phase and the beginning of the elimination phase.
  • TG-1101 serum concentration-time, dose, demographic, and covariate data from one Phase 2 study (TG1101-RMS201) and two Phase 3 studies (TG1101-RMS301 and TGI 101- RMS302) in subjects with RMS were pooled for the Pop PK analysis of TG-1101.
  • the dataset was combined with a previous dataset of TG-1101 in subjects with hematologic malignancies.
  • TG-1101 was administered as a single agent and compared with placebo to examine the level of B cell depletion by TG-1101 as well as determine the optimal dose and infusion time for TG-1101 in subjects with RMS. Based on the results of this study, a dose of 150 mg (infused over 4 hours) on Week 1 Day 1 followed by a dose of 450 mg (infused over one hour) on Week 3 Day 15 resulted in a median of >99% B cell depletion that was achieved on Week 4 and sustained until Week 24. The dosing regimen was well tolerated by the subjects with infusion-related reactions (Grade 1 and 2) being the commonly reported adverse event. Two Phase 3 studies in subjects with RMS have been completed.
  • TGI 101-RMS301 (aka ULTIMATE I) and TGI 101-RMS302 (ULTIMATE II), were randomized, double-blind, double-dummy, active-controlled studies of TG-1101 compared with oral teriflunomide to assess ARR, safety and tolerability in subjects with RMS.
  • the dataset for PopPK analysis included a total of 8672 PK samples collected from 931 subjects. Pre-dose samples accounted for 10.02% of PK samples and records with missing information or outliers (> 10 standard deviations from the mean TG-1101 concentration at the nominal dosing time) accounted for 0.20% and were excluded. Post-dose samples that were BLQ accounted for 3.47% of the data. Exclusions resulted in no quantifiable post-dose PK samples for 36 subjects. Consequently, the PK analysis dataset included 7485 PK samples from 895 subjects, of which 5624 PK samples were from 591 subjects with RMS.
  • PK parameter estimates are presented in Table 20.
  • TG-1101 CL was modestly increased by 14% in subjects that were ADA positive compared to those had no quantifiable ADA.
  • CL ranged from 22% lower to 48% higher compared to that for a typical subject with a body weight of 73 kg.
  • CL was reduced by a median of 12.5%.
  • Figure 12 shows the Goodness-of-Fit (GOF) diagnostics of the TG-1101 final model.
  • Figure 13 shows the pcVPC for the TG-1101 final PK model by study.
  • Figure 14 shows the forest plot of covariate effects on TG-1101 drug exposure.
  • the final PopPK model was re-estimated with the dataset including excluded outliers. All structural parameters were precisely estimated with no relevant change in those estimates, while the IIV terms were inflated with the inclusion of the outliers. It can therefore be expected that exclusion of outliers during the model development eliminated the influence of these outliers and minimized spurious findings in the covariate analysis.
  • the final PopPK model was also re- estimated including the RMS subpopulation only. The fit of the model to the RMS data resulted in similar structural parameters and random effects, with the exception of marginally lower IIV on CL; precision of parameter estimation was also similar.
  • TG-1101 The final model of TG-1101 was utilized to obtain individual post hoc estimates of PK parameters.
  • PK parameters AUCss, Cavg,ss, Cmax,ss, and Cmin,ss
  • the geometric mean / 2 (90% confidence interval [CI]) was calculated to be 21.8 days (21.4, 22.1 days). Median time to reach steady state was determined to be 15. 5 weeks. Accordingly, there was no accumulation for subjects that received the per- protocol regimen of 150 mg TG-1101 on Day 1 followed by 450 mg on Day 15, Week 24 and Week 48.
  • the median Cmax ratio at Week 24 to Cmax on Day 1 was 3.04 (range 3.00 to 3.42) consistent with the 3- fold increase in the amount of the dose and indicative of no accumulation. Similarly, the Cmax ratio at Week 48 to Week 24 was 1, indicative of no accumulation.
  • the model-predicted geometric mean AUCss, Cavgss, Cmaxss, and Cminss was 3000 ug d/mL ( ⁇ 28%), 8940 ng/mL ( ⁇ 28%), 139000 ( ⁇ 15.1%), and 139 ( ⁇ 170%), respectively.
  • TG-1101 began with the thaw of a working cell back (WCB) vial, described further below.
  • the culture was expanded through a series of shake flasks and seed bioreactors to meet the inoculum requirements of the 15,000 L production bioreactor, which was operated in fed-batch mode.
  • the bioreactor was harvested and clarified by centrifugation followed by depth filtration.
  • the clarified harvest was purified by three chromatography steps including Protein A, cation exchange, and anion exchange, designed to purify the TG-1101 and to reduce process impurities such as host cell protein and residual DNA.
  • the purification process (as illustrated in Example 4) contained steps to ensure viral safety including viral inactivation (solvent/detergent) and a viral filtration step.
  • Final UFDF and formulation steps are used to concentrate and buffer exchange TG-1101 into the formulation buffer and to the desired product concentration.
  • the ready to fill drug substance was formulated to obtain TG-1 lOlat a concentration of 25.0 mg/mL in 25 mM sodium citrate, 154 mM sodium chloride, 0.07% polysorbate 80, pH 6.5. After filling, the TG-1101 drug substance was frozen at ⁇ -60°C and then stored frozen at ⁇ -35 °C.
  • the host cell line used for generation of the TG-1101 producing cell line was the rat cell line YB2/0.
  • the production cell line, R603-12D11 was developed after transfecting the expression vector HK463-25 (containing the immunoglobulin heavy and light chain cDNA sequences of TG-1101) into the YB2/0 host cell line.
  • Figure 15 depicts the expression vector map of HK463-25 to produce TG-1101, in a 15,000 L bioreactor.
  • the expression vector HK463-25 included various elements that were optimized for stable expression in the YB2/0 host cell line.
  • the Rous Sarcoma Virus Long Terminal Repeat (RS V LTR) promoter was used for the constitutive expression of both heavy and light chain cDNAs. This promoter corresponds to the Long Terminal Repeat of the RSV genome which contains enhancer elements in its 5’ region and has strong transcriptional activity in the YB2/0 cell line.
  • Transcriptional termination and polyadenylation of both heavy and light chain cDNAs were provided by the human growth hormone polyadenylation sequence (hGH poly A).
  • hGH poly A human growth hormone polyadenylation sequence
  • a chimeric intron was introduced 5’ to the cDNA sequence of each antibody chain to improve expression.
  • This intron was optimized for splicing and is composed of a 5’ donor sequence from human beta-globin and a 3 ’ acceptor sequence from an Ig heavy chain variable gene.
  • the beta lactamase gene conferred ampicillin resistance (AmpR) and was provided to enable production of the plasmid in E. coli.
  • the enzyme neomycin-phosphotransferase II (NeoR) was under the control of the SV40 promoter and confers resistance to the transfected cell line to the antibiotic G418, thus acting as a selectable marker.
  • Dihydrofolate reductase was under the control of the SV40 promoter and conferred resistance to methotrexate (MTX) and can also act as a selectable and amplifiable marker in the transfected cell line.
  • MTX methotrexate
  • the HK463-25 expression vector (Figure 15) was 11.1 kb in size and contained five open reading frames for the antibody heavy chain, light chain, Dhfr, NeoR and AmpR genes in the same orientation.
  • the restriction sites shown in the figure were used for Southern blot analysis of the integration of the construct.
  • a unique Notl restriction site located 3’ to the NeoR gene was used for linearization of the vector prior to transfection.
  • the production cell line, R603-12D11 The production cell line, R603-12D11
  • the production cell line, R603-12D11 was generated after transfection of the host cell line, selection and screening of transfectants, and then limiting dilution cloning. The clones were screened, production cell line R603-12D11 was selected, and adapted to serum free medium. A pre-seed stock (PSS) cell bank was prepared. An overview of the steps involved in the generation of production cell line R603-12D11 is shown in Table 22.
  • a cryovial of the YB2/0 cell bank (YB2/0-301 04/147) was thawed and the cells were grown by dilution to a cell density of 1 * 10 5 cells/mL every 3 to 4 days with fresh culture medium (EMS medium with 5% FCS). The cells were seeded at a density of 2 10 5 cells/mL on the day before transfection in order to reach exponential phase prior to transfection. 44.5 pg of the NotLlinearized expression vector HK463-25 ( Figure 15) was transfected into 5/ 10 6 cells by electroporation using the animal component free Optimix reagent (Equibio). The cells were diluted in culture medium and seeded at 100 cells/well in 96-well plates. Selection with 1 g/L G418 in the culture medium was initiated three days after electroporation.
  • transformants were initially screened for titer by ELISA after selection in the G418 medium. Over 3000 transformants were screened and over 200 of the best producing wells were selected for further testing and for continued passaging. A second titer screen was performed to further reduce the number of clones. This screen was followed by additional screening for antibody fucose level by ELISA. Fucose levels less than 40% were considered desirable in order to provide the expected level of activation of CD 16 by the antibody; CD 16 activation is inversely related to the fucose levels.
  • the selected cell line was expanded from the cloning step, it also underwent a transition into serum free media and was re-screened to ensure the desired phenotype before the preparation of a small cell bank.
  • This small cell bank was then thawed and expanded to generate the R603-12D11 pre-seed stock cell bank (PSS).
  • PSS pre-seed stock cell bank
  • MCP Master Cell Bank
  • the MCB was manufactured at Henogen (later acquired by NovaSep).
  • the MCB lot G071/MCB/070208 was prepared by thawing and expanding one vial of the production cell line R603-12D11 pre-seed stock in serum-free medium EM-SF2 P500 H4 (EMS basal medium supplemented with 2-mercaptoethanol, ethanolamine, NaHCO, ferric citrate, pluronic acid, HEPES and recombinant human insulin).
  • the cells were expanded in T-flasks and roller bottles for eleven days.
  • the cell suspension was concentrated by centrifugation and aliquoted into 13 separate fractions.
  • each fraction was centrifuged and resuspended to a target cell density of 10* 10 6 cell/mL in freezing medium (90% EM-SF2 P500 H4 + 10% DMSO).
  • the suspended fractions were then each aliquoted into 18 cryovials per fraction resulting in a total of 234 cryovials of the MCB.
  • the cryovials were placed on dry ice and then into cryoboxes which were placed in a -80°C freezer for 21 hr.
  • the cryovials were transferred into liquid nitrogen tanks on 19 February 2007 for long-term storage and are currently stored in multiple locations.
  • the number of cell generations from the PSS of the production cell line to the MCB is 10.4.
  • Southern blot assessment of the plasmid integration site demonstrated comparable hybridization patterns for the MCB and PSS.
  • the number of integration sites for both the MCB and WCB was similarly measured to be 1 in both cell banks via Southern blot.
  • intra-chromosomic integration of the HK463-25 expression plasmid with insertion at a single locus of a metacentric chromosome was demonstrated by fluorescence in situ hybridization (FISH) analysis.
  • FISH fluorescence in situ hybridization
  • NGS Next generation nucleic acid sequencing
  • TLA targeted locus amplification
  • the first WCB was manufactured at NovaS ep (after acquiring Henogen). To prepare the WCB, one vial of MCB G071/MCB/070208 was thawed and expanded in serum-free medium EM-SF2 P500 H4 in flasks and roller bottles over eleven days. The expanded cell suspension was concentrated by centrifugation and aliquoted into 22 identical fractions. Each fraction was centrifuged and resuspended to a target cell density of 12.1 10 6 cell/mL in freezing medium (90% EM-SF2 P500 H4 + 10% DMSO). The suspended fractions were then each aliquoted into 18 cryovials resulting in a total of 396 cryovials of the WCB.
  • the lot was designated as G140/R603/WCB001.
  • the cryovials were placed on dry ice and then into cryoboxes which were placed in a -80°C freezer for 24 hr.
  • the cryovials were transferred into liquid nitrogen tanks for long-term storage on 22 September 2009.
  • the WCB is stored in at least two different storage sites including smaller numbers of vials stored for shorter duration at the manufacturer.
  • the number of cell generations from the PSS of the production cell line to the WCB is 21.4.
  • Testing of WCB lot G140/R603/WCB001 was performed including identity testing. Testing results confirmed the identity of the WCB as rat derived. Performance qualification was confirmed by thawing a vial of the WCB and monitoring for cell viability, doubling time, and IgG productivity. Assessments of copy number, restriction endonuclease profile, and number of integration sites, were conducted. The number of heavy and light chain copies integrated into the genome was estimated at 1.2 and 2.5, respectively, by quantitative polymerase chain reaction (Q-PCR). These results are consistent with that of the MCB. Southern blot assessment of the plasmid integration site demonstrated comparable hybridization patterns for the WCB and MCB. The number of integration sites for the WCB was measured to be 1 via Southern blot.
  • NGS Next generation nucleic acid sequencing
  • TLA targeted locus amplification
  • Table 23 Phenotypic Stability Results of MCB Lot G071/MCB/070208
  • Table 24 Phenotypic Stability Results of WCB Lot G140/R603/WCB001
  • a process flow diagram of the upstream unit operations including operational controls and in-process controls are provided in Table 26. Among other controls, bioburden and endotoxin were measured in the batched media of the seed and production bioreactor stages. Table 26: Upstream Operational and In-Process Controls
  • CDM4Mab Cell culture growth medium
  • feeds BalanCD CHO Feed4, glucose feed, and glutamine feed
  • WFI Water for Injection
  • Media and feeds were filtered ( ⁇ 0.2 pm) into sterile vessels and stored prior to use as needed.
  • a cholesterol lipid concentrate was supplemented into the CDM4Mab growth medium during preparation to support cell growth from the inoculum expansion stages through the production bioreactor.
  • the cholesterol lipid concentrate was added to the production bioreactor as fixed bolus feed additions on process days 0 and 4. Operational and in-process controls for cell culture media and feeds are described in Table 27.
  • Table 27 Cell Culture Media and Feeds Preparation and Storage Controls a Room temperature at SBL: 17-25°C b Operating temperature: 37.0°C (36.5-37.5°C)
  • Inoculum expansion steps included thaw of the WCB vial and growth in shake flasks and/or cellbags of increasing size and volume to provide sufficient cell mass to inoculate the seed bioreactor stages.
  • the steps are performed by growth in inoculum expansion growth medium (CDM4Mab).
  • CDM4Mab inoculum expansion growth medium
  • a vial of WCB G140/R603/WCB001 was thawed in pre- warmed water in a 37.0°C water bath.
  • the thawed vial contents were transferred into prewarmed medium and diluted to achieve a target seed density of 0.55 x 10 6 viable cells/mL.
  • the culture was placed in an initial 125 mL shake flask and grown in a shaking incubator at 37.0°C/5.0% CO2 for 1 day. Every 2 to 3 days, the culture was expanded into shake flasks of larger volume and/or to multiple shake flasks. At each stage, a seeding density of 0.30 x 10 6 viable cells/mL was targeted. The final inoculum preparation stage consisted of a 50 L cellbag. After 2 to 3 days of growth, the viable cell density was checked and culture was further processed to the seed bioreactor stages. Operational and in-process controls for inoculum expansion are described in Table 28.
  • Table 28 Inoculum Expansion Controls a Shaker platform throw radius dependent parameter. Shaker platform throw radius: 22 mm
  • the seed bioreactor stages further increase the volume and cell culture biomass prior to inoculation of the production bioreactor.
  • the medium used in these stages was inoculum expansion growth medium (CDM4Mab).
  • the seed bioreactor stages were 120 L, 600 L, and 3000 L stainless steel bioreactors.
  • the bioreactors were equilibrated after media addition. Dissolved oxygen and pH probes were calibrated prior to use.
  • Initial bioreactor set points included temperature, pH, dissolved oxygen, and agitation rate.
  • Each bioreactor was inoculated with cell culture from the preceding stage and the culture was grown for 2 to 3 days. Seed bioreactor operational and in-process controls are summarized in Table 29.
  • the production bioreactor stage is the final cell culture process stage which further increases the volume and mass of the cell culture for expression of the TG-1101 antibody with acceptable product quality.
  • the basal medium used in this stage was growth medium (CDM4Mab) and feed additions were performed at specified days or criteria during the process.
  • the production bioreactor was a 15,000 L stainless steel bioreactor.
  • the production bioreactor was equilibrated after media addition. Inoculum expansion medium was added to an initial volume target. Dissolved oxygen and pH probes were calibrated prior to use. Initial bioreactor set points included temperature, pH, dissolved oxygen, and agitation rate. The bioreactor was inoculated with cell culture from the N-l seed bioreactor at a viable cell density target of 0.5 x 10 6 viable cells/mL. During the production bioreactor process, pH was controlled with base addition and CO2 sparging on demand. Dissolved oxygen was controlled with oxygen and air sparging on demand. Antifoam was added as needed to mitigate foaming concerns.
  • Offline pH, pCO2, pO2, osmolality, and metabolites were monitored daily as were viable cell density (VCD) and viability.
  • the BalanCD CHO Feed4 was added on Days 3, 5, 7, 9 at specified volumes (4.0% of initial bioreactor working volume).
  • a calculated volume of glucose solution was fed (to 4.00 g/L) when daily bioreactor sample measured values were ⁇ 3.00 g/L on specified process days.
  • a fixed volume of glutamine solution was bolus fed on Day 3 (3.0% of initial bioreactor working volume), and a calculated volume was fed (to 4.00 mM) when daily bioreactor sample measured values were ⁇ 3.00 mM on specified process days.
  • the production bioreactor was harvested based on either culture duration or cell viability criteria, whichever occured first. [00510] Unprocessed bulk samples were removed prior to the clarification unit operation for adventitious agent testing as described in Example 4. Production bioreactor operational and in- process controls are described below in Table 30.
  • Table 30 Production Bioreactor Controls a Temporary excursions are allowed. b Agitation is dependent on volumetric scale and bioreactor impeller. Target (NOR) are equivalent to power/volume 50 (42-59) W/m3 at SBL. AR is equivalent to power/volume 42-70 W/m 3 . c Fed on Days 3, 5, 7, 9. d Fed on Days 4-7 as needed. e Glucose/glutamine are not measured after glucose/glutamine feed addition; values refer to target concentrations used in the calculation for determining the glucose/glutamine feed amount required. f Fed on Days 3-12 as needed. g Antifoam amount is calculated based on initial working volume of production bioreactor. Target is addition amount at one time.
  • h Harvest occurs when either culture duration reaches its target/NOR, or when final cell viability drops lower than its NOR, whichever occurs first. As a result, it is possible for harvest to occur when final cell viability is below its NOR, or culture duration is below its NOR.
  • a process flow diagram of the downstream steps including operational controls and in-process controls are provided in Table 31. As shown in Table 31, in-process controls were incorporated into the process. Among other controls, bioburden and endotoxin were measured during multiple stages of the downstream process. TABLE 31: Downstream Operational and In-Process Controls
  • the cell culture supernatant was harvested and clarified from the 15,000 L bioreactor to remove cells and cell debris.
  • the clarification was performed using a continuous centrifugation followed by depth filtration.
  • the harvest clarification step was operated in a room in which the controlled temperature range was 17-25°C; the harvest pool vessel was a jacketed tank, which maintains the pool at 2-8°C.
  • the centrifuge shot interval was set based on the packed cell volume percentage (PCV) and adjusted to allow for 80% bowl fill.
  • PCV packed cell volume percentage
  • the flow rate to the centrifuge was actively controlled; the filtration feed flow was the same as the centrifuge feed flow.
  • Process parameters included centrate back pressure, the depth filtration operating pressure, inlet pressure, harvest weight, bioburden and endotoxin.
  • the centrate was clarified by use of a three-stage filtration process as required to pass through a 0.2 pm filter (Millistak A1HC POD depth filters, 1.2/0.5 pm filters and 0.45/0.22 pm sterilizing grade filters). Before use, the filters were flushed with WFI followed by equilibration. The centrate was pumped through the filters, which are monitored to ensure acceptable back pressure. Air was used to expel the contents in the filters followed by a buffer flush. The clarified harvest was stored at 2 to 8°C for ⁇ 11 days.
  • Protein A column chromatography was performed using MabSuRe Select resin (Cytiva) in bind/elute mode. This step provided capture and purification of TG-1101 with reduction of process impurities such as cell culture components, HCP, and residual DNA, as well as for the provision of viral safety.
  • the packed column was assessed for HETP performance using sodium acetate/benzyl alcohol buffer. During manufacturing, all column operations were performed at 13 to 25°C. Before loading, the column was sanitized with 0.5 M sodium hydroxide and flushed with WFI. The column was equilibrated with Equilibration Buffer (25 mM Tris, 25 mM NaCl, 5 mM EDTA, pH 7.1).
  • the eluate was collected in a tank containing neutralization buffer (2.0M Tris, pH 7.5) and filtered via a 0.2 pm filter before transfer to a different tank.
  • neutralization buffer 2.0M Tris, pH 7.5
  • the Protein A column was sanitized with 0.5M sodium hydroxide. Up to three cycles per batch may be run; if multiple cycles were required for the Protein A process, the column was re-equilibrated for the next cycle with equilibration buffer. After sanitization, the Protein A column was neutralized with equilibration buffer and stored at 13 to 25°C in 200 mM sodium acetate, 2% benzyl alcohol, pH 5.0.
  • the pooled (if more than one cycle), neutralized eluate was diluted with 5mM Sodium Phosphate, pH 7.2 to a concentration of ⁇ 10 g/L and stored at 13-25°C for ⁇ 24 hours or at 2 to 8°C for ⁇ 11 days.
  • the Protein A capture chromatography step was followed by a solvent detergent viral inactivation (SDVI) step to inactivate potential viral agents.
  • SDVI solvent detergent viral inactivation
  • the Protein A elution pool was diluted and treated with 3.5% (v/v) TnBP, 12% (w/v) polysorbate 80 and held at 24.0-26.0°C for at least 120 minutes while mixing.
  • the SDVI pool was filtered with a 0.2 pm filter before transfer to a different tank, where it was diluted with 5 mM sodium phosphate, pH 7.2 to 50 mOsm/kg, and the pH adjusted to 7.2 as needed. After pH adjustment, the pool was held at 13 to 25°C for ⁇ 30 hours before proceeding to the CEX column.
  • Cation Exchange Chromatography (CEX) Cation Exchange Chromatography
  • Cation exchange column chromatography was performed using SP Sepharose Fast Flow (Cytiva) in bind/elute mode. This step provided further purification of TG-1101, removing residual process impurities (HCPs, DNA, residual polysorbate 80, and TnBP).
  • the packed column was assessed for HETP performance using a sodium acetate/benzyl alcohol-containing buffer. During manufacturing, all column operations were performed at 13 to 25°C. In the currently validated manufacturing process, before loading, the column was sanitized with 0.5 M sodium hydroxide and rinsed with WFI. The column was equilibrated using equilibration buffer (20 mM sodium phosphate, pH 7.2). The viral inactivated/diluted solution was loaded onto the column at a maximum of 65 g/L resin load. The column was washed with Wash 1 Buffer (equilibration buffer) followed by a second wash with Wash 2 Buffer (equilibration buffer in the reverse direction).
  • Wash 1 Buffer equilibration buffer
  • Wash 2 Buffer equilibration buffer in the reverse direction
  • Bound TG-1101 was eluted using 20 mM sodium phosphate, 150 mM NaCl, pH 7.2 and elution peak collection by A280 monitoring. The eluate was filtered (0.2 pm) and stored at 13 to 25°C for ⁇ 72hours or at 2 to 8°C for ⁇ 11 days. After elution, the column was stripped with 2 M NaCl followed by sanitization with 0.5 M sodium hydroxide. One cycle per batch was allowed. After completion, the column was sanitized (0.5 M sodium hydroxide) and stored in storage buffer (200 mM sodium acetate, 2% benzyl alcohol, pH 5.0).
  • Anion exchange membrane chromatography was performed using a Mustang Q (Pall Corporation) membrane absorber (MA) filter in flow-through mode. This step provided further purification of the TG-1101; the product flowed through the membrane and remaining impurities (DNA, HCP and viruses) were retained on the membrane.
  • the membrane was single use (i.e., each individual membrane cannot be reused) and several membrane capsules may be used per batch at the appropriate loading level.
  • eluate from the cation exchange column chromatography step was diluted with 20 mM sodium phosphate, pH 8.0, followed by the adjustment of the pH to 8.0.
  • concentration was determined and the number of cycles calculated based on protein concentration such that the loading was 200 to 700 g TG-1101/L membrane load.
  • the membrane was sanitized with 0.5 M sodium hydroxide, flushed with 2 M NaCl then with WFI, before being equilibrated with Equilibration Buffer (20 mM sodium phosphate, 75 mM NaCl, pH 8.0) in preparation for the load.
  • the membrane was chased with 20 mM sodium phosphate, 75 mM NaCl, pH 8.0, to maximize recovery.
  • the collected flow- through containing product from all cycles was filtered (0.5/0.2 pm), diluted to ⁇ 6 g/L with 75 mM sodium citrate, 312 mM NaCl, pH 6.0, and the pH is adjusted to 6.8.
  • the adjusted AEX pool was stored at 13 to 25°C for ⁇ 72 hours or at 2 to 8°C for ⁇ 11 days.
  • Viral filtration was performed by filtering through a Viresolve prefilter in series with a Viresolve Pro viral filter (Millipore Sigma), at an operating temperature Target Range of 13- 25°C.
  • the process used sufficient filters to meet the loading limit.
  • the step was designed to remove potential viruses, including small viruses such as parvovirus.
  • the filters were set up in series and flushed with WFI, integrity tested, then sanitized with 0.5 M sodium hydroxide. This was followed by flushing with equilibration buffer (25 mM sodium citrate, 154 mM NaCl, pH 6.5). The filtered Mustang Q membrane flow-through was processed through the viral reduction filters.
  • Protein concentration was determined and used to confirm the membrane load ratio was ⁇ 600 g/m 2 After loading, the membrane was chased with equilibration buffer and post-use integrity testing was performed. Viral filtrate was stored at 13 to 25°C for ⁇ 72 hours or at 2 to 8°C for ⁇ 11 days.
  • the UFDF step was used to concentrate the viral filtrate and buffer exchange into diafiltration buffer at an operating temperature Target Range of 13-25°C.
  • the process used a tangential flow filter with a 30 kDa molecular weight cut-off.
  • the setup included up to sufficient filters to meet the loading limit of ⁇ 250 g/m 2 .
  • the membranes were sanitized with 0.5M NaOH, flushed with WFI, and flushed/equilibrated with diafiltration buffer before the start of the unit operation.
  • the unit operation included an initial ultrafiltration (UF1) step in which the TG-1101 was first concentrated to a target of 39 mg/mL. This was followed by diafiltration (DF) into diafiltration buffer (25 mM sodium citrate 154 mM NaCl, pH 6.5) with 8 diavolumes with an upper limit of ⁇ 9.2 diavolumes.
  • the ultrafiltration system was flushed with diafiltration buffer to maximize recovery; the flush was transferred to the formulation vessel and combined with the UF/DF Pool.
  • the membranes were flushed with WFI, cleaned with 0.5M NaOH, 250 ppm sodium hypoclorite, flushed with WFI, and stored in 0.1 M sodium hydroxide.
  • the diluted UFDF pool was stored at 15 to 25°C for ⁇ 18 hours.
  • the formulation step included the addition of concentrated polysorbate 80 in formulation buffer to achieve the final drug substance formulation, at an operating temperature Target Range of 17-25°C.
  • the step was performed by adding the stabilization buffer (25 mM sodium citrate 154 mM NaCl, 10 g/L polysorbate 80, pH 6.5).
  • the pool was diluted to a target of 23.5 to 26.5 mg/mL with 25 mM sodium citrate 154 mM NaCl, 700 mg/L polysorbate 80, pH 6.5, resulting in a ready-to-fill drug substance in the formulation buffer of 25 mM sodium citrate, 154 mM NaCl, 0.07% polysorbate 80, pH 6.5.
  • the formulated bulk drug substance was transferred and 0.2 pm filtered into 6 L Celsius® FFT bags in a closed, single use system to a target fill volume of 5.50 L. After filling, the formulated bulk drug substance was frozen at ⁇ -60 for >17 hours and stored at ⁇ -35°C.
  • This Example describes evaluation of the timing of onset of no evidence of disease activity (NEDA) and proportion of participants maintaining NEDA with ublituximab in pooled post hoc analyses of ULTIMATE I and II.
  • ULTIMATE I NCT03277261
  • ULTIMATE II NCT03277248
  • RMS relapsing multiple sclerosis
  • Ublituximab is glycoengineered for enhanced antibody-dependent cellular cytotoxicity and is administered in 1-hour infusions after the first infusion.
  • ublituximab significantly improved annualized relapse rate as well as number of gadolinium-enhancing (Gd+) T1 lesions and new/enlarging T2 lesions, and a higher proportion of participants achieved 3 -parameter NEDA (NEDA-3) rates with ublituximab vs teriflunomide.
  • NEDA rates were 44.6% vs 12.4% at Weeks 0-96 for ublituximab vs teriflunomide cohorts, respectively, and 82.1% vs 22.5% at Weeks 24-96 (rebaselined; PO.OOOl for both).
  • Weeks 24-96 re-baselined
  • 17.9% of ublituximab- treated participants had evidence of disease activity (EDA), and relapse was the most common component (11.4% ublituximab vs 22.9% with teriflunomide).
  • EDA disease activity
  • 77.5% of teriflunomide-treated participants had EDA, and the most common component was new/enlarging T2 lesions (71.6% with teriflunomide vs 3.1% with ublituximab).
  • MRI magnetic resonance imaging
  • NEDA e.g., NEDA-3
  • NEDA-3 was defined as no confirmed relapses, no Gd+ T1 lesions, no new/enlarging T2 lesions, and no 12- week confirmed disability progression. The results are described in Figures 16A-16C.
  • This Example describes characterization of cytopenias with ublituximab in the ULTIMATE I and II studies.
  • pharmacodynamic (PD) studies of ublituximab have reported a transient decline in the percentage of total T cells and NK cells as well as a reciprocal increase in myeloid cells following the initial dose of ublituximab at Day 2 (Fox EJ, et al. Presented at: ACTRIMS (February 24-26, 2022), West Palm Beach, FL, Poster P105; Lovett-Racke AE, et al. J Neuroimmunol. 2021, 359:577676).
  • Lymphocyte counts at Day 2 have not been evaluated in pivotal trials of other anti-CD20 agents (Hauser SL, et al. N Engl J Med. 2017, 376(3):221-234; Hauser SL, et al. N Engl J Med. 2020, 383(6): 546-557). Additional evaluations of ULTIMATE I and II data were conducted to understand the incidence and kinetics of cytopenias with ublituximab as well as potential association with infections. In the Phase 3 ULTIMATE I and II studies in participants with RMS), adverse events with ublituximab overall were not imbalanced compared with teriflunomide and were consistent with the anti-CD20 class.
  • the ULTIMATE I and II enrolled a total of 1094 adults from 10 countries with a diagnosis of RMS (relapsing-remitting or secondary-progressive) with disease activity (Steinman L, et al. N Engl J Med. 2022, 387(8):704-714).
  • cytopenia as a TEAE (any grade/Grade >3) was reported in 25.7%/12.1% of ublituximab participants.
  • the most commonly reported cytopenias (all Grade/Grade >3) were lymphopenia (18.7%/9.5%), neutropenia (3.3%/2.0%), leukopenia (3.3%/0.2%), and anemia (2.9%/0.0%).
  • Example 18 Phase 3 Results of Ublituximab compared to Teriflunomide in Treating Patients with Relapsing Forms of Multiple Sclerosis (RMS) - Annualized Relapse Rate (ARR)
  • ULTIMATE I and ULTIMATE II two identical, independent, phase 3 randomized, multi-center, double-blinded, active-controlled clinical trials, conducted in parallel, enrolled 1094 patients with relapsing forms of multple sclerosis (RMS) across 10 countries, to evaluate the safety and efficacy of the glycoengineered anti-CD20 antibody ubliximab versus teriflunomide in these patients.
  • RMS multple sclerosis
  • the primary endpoint for the ULTIMATE I and ULTIMATE II phase 3 clinical studies was the Annualized Relapse Rate (ARR) following 96 weeks of treatment.
  • ARR refers to the ratio of the sum of the subject’s RMS relapse counts divided by the sum of the subject’s treatment duration (in years).
  • Key secondary endpoints for ULTIMATE I and ULTIMATE II included total number of gadolinium-enhancing T1 lesions on MRI by Week 96; total number of new and/or enlarging T2 hyperintense lesions by Week 96; and the proportion of subjects with No Evidence of Disease Activity (NED A) from Week 24 to Week 96.
  • CDP Confirmed Disability Progression
  • CDI Confirmed Disability Improvement
  • the primary endpoint for the ULTIMATE I and ULTIMATE II phase 3 clinical studies was Annualized Relapse Rate (ARR) following 96 weeks of treatment. Both the ULTIMATE I and ULTIMATE II studies met their primary endpoint with ublituximab treatment demonstrating a statistically significant reduction in annualized relapse rate (ARR) over a 96- week period (p ⁇ 0.005 in each trial). Ublituximab treatment resulted in an ARR of ⁇ 0.10 in each of ULTIMATE I and ULTIMATE II.
  • T1 hypointense lesions on MRI were evaluated.
  • T1 “black holes” are hypointense lesions commonly seen on T1 -weighted images in patients with multiple sclerosis, and are indicative of chronic stage disease associated with white matter destruction, axonal loss, and irreversible clinical outcome. See, Kocsis, K. el al., Frontiers in Neurology 12'. 1-8 (March 2021).
  • T1 hypointense lesions were a tertiary endpoint in the individual studies. Pooled analyses of T1 hypointense lesions were performed post hoc. T1 hypointense lesion volume was assessed using mixed model repeated measures of the change in cubic root transformed volume from baseline at scheduled visits up to 96 weeks.
  • the mean volume of T1 hypointense lesions (mL) at baseline, Week 24, Week 48, and Week 96 was 3.280, 3.266, 3.244, and 3.132 for ublituximab and 3.343, 3.360, 3.330, and 3.475 for teriflunomide, respectively.
  • the mean number of new T1 hypointense lesions at 96 weeks was significantly reduced with ublituximab versus teriflunomide (1.5 ⁇ 3.55 vs 5.4 ⁇ 10.67; P ⁇ 0.0001).
  • PK Pharmacokinetics
  • PK pharmacokinetics

Abstract

Provided herein are populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications. Also provided are methods of using and methods of making such populations of anti-CD20 antibody proteins.

Description

ANTLCD20 ANTIBODY COMPOSITIONS
1. CLAIM OF PRIORITY
[0001] This application claims the benefit of U.S. Provisional Application Serial No. 63/347,852, filed on June 1, 2022, U.S. Provisional Application Serial No. 63/421,078, filed on October 31, 2022, and U.S. Provisional Application Serial No. 63/445,082, filed on February 13, 2023. The entire contents of the foregoing are hereby incorporated by reference.
2. REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[0002] This application contains a Sequence Listing that has been submitted electronically as an XML file named “50581 -0004W01. XML.” The XML file, created on May 22, 2023, is 39,114 bytes in size. The material in the XML file is hereby incorporated by reference in its entirety.
3. FIELD OF THE DISCLOSURE
[0003] The present disclosure is in the field of recombinant anti-CD20 antibodies, methods of producing such antibodies, and uses of such antibodies.
4. BACKGROUND OF THE DISCLOSURE
[0004] Therapeutic monoclonal antibodies (mAbs) produced in mammalian cells are heterogeneous as a result of post-translational modifications (PTMs). PTMs can occur during mAb production, purification, storage, and post-administration. PTMs are therapeutic mAb product quality attributes (PQAs). Controlling PQAs within predefined acceptance criteria is vital to the biopharmaceutical industry because it ensures consistent product quality and reduces potential impacts on drug safety and efficacy (Xu, X. el al., Journal of Applied Bioanalysis 3(2):21-5 (2017)).
[0005] The critical importance of sequence variation in antibodies is well recognized. Sequence diversity in antibody variable domains is essential for specific antigen recognition while linkage to different constant domains leads to distinct Fc-mediated effector activities. PTMs of these domains provide an additional immune mechanism by which the binding and activity of antibodies can be modulated. PTMs vary from chain additions, such as N- and O- linked glycosylation, glycation, cysteinylation and sulfation; chain trimming, such as C-terminal lysine clipping; amino acid modifications such as cyclization (into a N-terminal pyroglutamic acid), deamidation, oxidation, isomerization and carbamylation; to disulfide scrambling of hinge region interchain disulfide bonds. Each antibody can therefore give rise to a myriad of distinct antibody molecules with large activity and potency differences. Although post-translational modifications of antibodies have been observed and studied for decades, the full impact of the microheterogeneity is yet to be further studied. PTMs can impact antibody functions, for example, pharmacokinetics and pharmacodynamics properties and clinical efficacy.
[0006] Having specified ranges of post-translational modifications can be critical for a population of mAbs to possess consistent product quality, clinical safety, and efficacy.
5. SUMMARY OF THE DISCLOSURE
[0007] Provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising about 20 to 40% fucosylated glycans and optionally about 10 to 20% galactosylated glycans.
[0008] In some embodiments, the N-glycan profile comprises 23% to 36% fucosylated glycans, optionally about 30% fucosylated glycans. In some embodiments, the N-glycan profile comprises 16% to 18% galactosylated glycans, optionally about 17% galactosylated glycans.
[0009] In some embodiments, the relative abundance of fucosylated glycans is the percent of fucosylated glycans among all glycans in the N-glycan profile. In some embodiments, the relative abundance of galactosylated glycans is the percent of galactosylated glycans among all glycans in the N-glycan profile.
[0010] In some embodiments, the N-glycan profile comprises 12% to 30% bisecting N- glycans, optionally about 18% bisecting N-glycans. In some embodiments, the bisecting N- glycans comprise one or more of GOB, G0FB, G1FB, G2FBS1, and G2FBS2.
In some embodiments, the population of anti-CD20 antibody proteins has an N-glycan profile comprising less than 5% sialylated glycans. In some embodiments, the N-glycan profile comprises less than 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycan. In some embodiments, the N-glycan profile comprises no detectable amount of sialylated glycan. [0011] In some embodiments, the population of anti-CD20 antibody proteins has an N- glycan profile comprising 0.1% to 1.5% Man5 N-glycan. In some embodiments, the N-glycan profile comprises 0.4% to 0.7% Man5 N-glycan. In some embodiments, the N-glycan profile comprises about 0.6% Man5 N-glycan. In some embodiments, Man5 N-glycan is the only high mannose species in the N-glycan profile.
[0012] In some embodiments, the population of anti-CD20 antibody proteins comprises 0.20 to 0.40 mol isoaspartate per mol protein. In some embodiments, the population of anti-CD20 antibody proteins comprises 0.25 to 0.35 mol isoaspartate per mol protein.
[0013] In some embodiments, glutamate at position 1 of the heavy chain is a pyroglutamate and glutamate at position 1 of the light chain is a pyroglutamate.
[0014] In some embodiments, the population of anti-CD20 antibody proteins has an N- glycan profile comprising a relative abundance ratio of 0.1 to 0.15 G1 to GO N-glycans. In some embodiments, the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.5 to 0.9 GIF to G1 N-glycans.
[0015] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance ranges:
(a) 0.3% to 2% G0-GN;
(b) 0.1% to 2% G0F-GN;
(c) 30% to 60% GO;
(d) 0.1% to 1% G1-GN;
(e) 5% to 20% GOB;
(f) 5% to 30% G0F;
(g) 0.1% to 1.5% Man5;
(h) l% to 15% G0FB;
(i) l% to 13% Gl;
(j) 0.5% to 10% Gl’;
(k) 0.5% to 6% GIB;
(l) 0.5% to 12% GIF;
(m) 0.1% to 3% GIF’;
(n) 0.1% to 3% G1FB; (o) 0.1% to 2% G2; and
(p) 0.1% to 2% G2F.
[0016] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
(a) 0.8% to 1.1% GO-GN;
(b) 0.5% to 1.1% G0F-GN;
(c) 42.5% to 48.8% GO;
(d) 0.3% to 0.6% Gl-GN;
(e) 9.5% to 14.1% GOB;
(f) 12.8% to 19.7% G0F;
(g) 0.4% to 0.7% Man5;
(h) 5.1% to 7.0% G0FB;
(i) 5.7% to 6.4% Gl;
(j) 2.7% to 3.3% Gl’;
(k) 1.4% to 2.0% GIB;
(l) 2.6% to 4.2% GIF;
(m) 1.1% to 1.6% GIF’;
(n) 1.1% to 1.8% G1FB;
(o) 0.5% to 0.7% G2; and
(p) 0.3% to 0.5% G2F.
[0017] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
(a) 0.9% G0-GN;
(b) 0.8% G0F-GN;
(c) 46.1% GO;
(d) 0.5% Gl-GN;
(e) 10.9% GOB;
(f) 17.0% G0F;
(g) 0.6% Man5;
(h) 6.0% G0FB;
(i) 6.1% Gl; (j) 2.9% Gl’;
(k) 1.6% GIB;
(l) 3.2% GIF;
(m) 1.3% GIF’;
(n) 1.3 G1FB;
(o) 0.5% G2; and
(p) 0.3% G2F.
[0018] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least three, four or five N-glycans within the following relative abundance ranges:
(a) 0.3% to 2% G0-GN;
(b) 0.1% to 2% G0F-GN;
(c) 30% to 60% GO;
(d) 0.1% to 1% G1-GN;
(e) 5% to 20% GOB;
(f) 5% to 30% G0F;
(g) 0.1% to 1.5% Man5;
(h) l% to 15% G0FB;
(i) l% to 13% Gl;
(j) 0.5% to 10% Gl’;
(k) 0.5% to 6% GIB;
(l) 0.5% to 12% GIF;
(m) 0.1% to 3% GIF’;
(n) 0.1% to 3% G1FB;
(o) 0.1% to 2% G2; and
(p) 0.1% to 2% G2F.
[0019] In some embodiments, the N-glycan profile of the population of anti-CD20 antibody proteins is determined using a method comprising: (a) incubating the population of anti-CD20 antibody proteins with an enzyme, wherein the enzyme catalyzes releasing of the N-glycans from the anti-CD20 antibody; (b) measuring the relevant abundance of the released N-gylcans using one or more methods selected from chromatography, mass spectrometry, capillary electrophoresis, and the combination thereof. In some embodiments, the method further comprises after step (a) and before step (b) the following steps: (c) purifying the N-glycans; and (d) labeling the N-glycans with a fluorescent compound. In some embodiments, the enzyme is PNGase F. In some embodiments, the fluorescent compound is 2-aminobenzamide (2- AB). [0020] In some embodiments, less than 10% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 5% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 1% of the anti- CD20 antibody proteins in the population is non-glycosylated.
[0021] In some embodiments, the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) 8.0% to 10.0% a-helix;
(b) 32.0% to 36.0% Anti-parallel P-sheet;
(c) 5.0% to 6.0% Parallel P-sheet;
(d) 16.0% to 18.0% P-Turn; and
(e) 35.0% to 36.0% random coil.
[0022] In some embodiments, the population of anti-CD20 antibody proteins comprises secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) 8.0% to 10.0% a-helix;
(b) 32.0% to 36.0% Anti-parallel P-sheet;
(c) 5.0% to 6.0% Parallel P-sheet;
(d) 16.0% to 18.0% P-Turn; and
(e) 35.0% to 36.0% random coil.
[0023] In some embodiments, the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) about 9.0% a-helix;
(b) about 33.0% Anti-parallel P-sheet;
(c) about 5.6% Parallel P-sheet;
(d) about 17.5% P-Turn; and
(e) about 35.2% random coil. [0024] In some embodiments, the population of anti-CD20 antibody proteins further comprises one or more of the following post-translational modifications at the specified abundance:
Figure imgf000009_0001
[0025] In some embodiments, the one or more of the post-translational modifications are measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS).
[0026] In some embodiments, the population has an amount of total protein of 25.5-25.8 mg/mL as measured by absorbance at 280 nm.
[0027] In some embodiments, the anti-CD20 antibody proteins in the population induces greater cytotoxicity in a cell-based antibody-dependent cellular cytotoxicity (ADCC) assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
[0028] In some embodiments, the population has a relative potency of 90 to 163% in a cellbased ADCC assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of 78% to 116% or 73% to 128% in a cell-based complement dependent cytotoxicity (CDC) assay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of 92 to 118% or 82 to 138% in a cellbased CD20 binding activity bioassay compared to that of a commercial reference standard. In some embodiments, the population has a KD value 30 to 70 nM in an FcyRIIIa-158V binding assay as measured by surface plasmon resonance. In some embodiments, the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In some embodiments, the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab. In some embodiments, the population has a relative potency of 88 to 113% or 86 to 116% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard. In some embodiments, the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard.
[0029] In some embodiments, the population has 99.2 to 99.9% monomers as detected by size exclusion chromatography (SEC). In some embodiments, the population has 0.1 to 0.8% dimers as detected by SEC. In some embodiments, the population has undetectable level of aggregates as detected by SEC; and/or undetectable level of fragments as detected by SEC. [0030] In some embodiments, the population has 93.6 to 95.9% IgG after purification by non-reduced capillary gel electrophoresis (CGE). In some embodiments, the population has 0.1 to 0.3% high molecular weight species (HMWS) after purification by non-reduced CGE. In some embodiments, the population has 0.7 to 1.2% free light chain (LC) after purification by non-reduced CGE. In some embodiments, the population has 97.7 to 98.0% heavy chain plus light chain species (HC + LC) after purification by reduced CGE.
[0031] In some embodiments, the population has 20 to 25% acidic isoforms as detected by imaged capillary isoelectric focusing (iCIEF). In some embodiments, the population has 50 to 60% main isoforms as detected by iCIEF. In some embodiments, the population has 20 to 30% basic isoforms as detected by iCIEF. In some embodiments, the population has an average molar ratio of free thiol to anti-CD20 antibody of about 2.0 to 2.2.
[0032] In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of the N-terminal residue. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 5 N- terminal residues. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 10 N-terminal residues. In some embodiments, terminal lysine amino acid residue of the heavy chain in the anti-CD20 antibody in the population is truncated.
[0033] Also provided here are pharmaceutical formulations comprising the compositions described herein, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 10 mg/mL to 50 mg/mL. In some embodiments, the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL.
[0034] In some embodiments, the pharmaceutical formulation further comprises one or more of the following: sodium chloride, trisodium citrate dehydrate, polysorbate 80, and hydrochloric acid. In some embodiments, the pharmaceutical formulation comprises about 9.0 mg/mL of sodium chloride, about 7.4 mg/mL of trisodium citrate dehydrate, about 0.7 mg/mL of polysorbate 80, and/or about 0.4 mg/mL of hydrochloric acid.
[0035] In some embodiments, the anti-CD20 antibody is present in a single dosage form.
[0036] Also provided here are pharmaceutical formulations comprising:
(i) the compositions described herein, wherein the composition comprises a single dosage form of the population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL,
(ii) about 9.0 mg/mL of sodium chloride,
(iii) about 7.4 mg/mL of trisodium citrate dehydrate,
(iv) about 0.7 mg/mL of polysorbate 80, and
(v) about 0.4 mg/mL of hydrochloric acid.
[0037] Also provided here are single batch preparations of populations of anti-CD20 antibody proteins or pharmaceutical formulations described herein, wherein the single batch comprises at least 100 g, at least 120 g, or at least 150 g of the anti-CD20 antibody proteins. [0038] Also provided here are populations of anti-CD20 antibody proteins or pharmaceutical formulations described herein, produced in a 15,000 L or 20,000 L bioreactor.
[0039] Also provided here are methods of treating multiple sclerosis (MS) in a subject in need thereof by administering to the subject a therapeutically effective amount of compositions or pharmaceutical formulations described herein.
[0040] In some embodiments, the composition or the pharmaceutical formulation is administered as i) a first infusion at a dose of about 150 mg of the the anti-CD20 antibody protein, ii) a second infusion two weeks later at a dose of about 450 mg of the the anti-CD20 antibody protein, and iii) subsequent infusions every 24 weeks or six months at a dose of about 450 mg of the the anti-CD20 antibody protein.
[0041] In some embodiments, administration of the composition or the pharmaceutical formulation to the subject results in one or more of the following pharmacokinetic parameters:
(a) an AUC between 2,160 pg/mL and 3,840 pg/mL;
(b) a Cmax between 118,011 ng/mL and 159,989 ng/mL;
(c) a Cmin between 40 ng/mL and 375 ng/mL; and
(d) a Cavg is between 6,437 ng/mL and 11,443 ng/mL.
[0042] In some embodiments, administration of the composition or the pharmaceutical formulation to the subject results in one or more of the following pharmacokinetic parameters:
(a) an AUC about 3,000 pg/mL;
(b) a Cmax about 139,000 ng/mL;
(c) a Cmin about 139 ng/mL; and
(d) a Cavg about 8,940 ng/mL.
[0043] In some embodiments, the method comprises a treatment period of at least 96 weeks.
[0044] In some embodiments, the subject has been pre-medicated with a corticosteroid 30-60 minutes prior to administration of the composition or the pharmaceutical formulation. In some embodiments, the corticosteroid is methylprednisone or dexamethasone. In some embodiments, the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
[0045] In some embodiments, the subject has been pre-medicated with an antihistamine 30- 60 minutes prior to administration of the composition or the pharmaceutical formulation. In some embodiments, the antihistamine is diphenhydramine HC1. In some embodiments, the diphenhydramine HC1 is administered at a dose of about 25-50 mg.
[0046] In some embodiments, the subject has been pre-medicated with an antipyretic 30-60 minutes prior to administration of the composition or the pharmaceutical formulation. In some embodiments, the antipyretic is acetaminophen or an antipyretic bioequivalent thereto.
[0047] In some embodiments, the subject has an Expanded Disability Status Scale (EDSS) score of from 0 to 5.5 prior to treatment.
[0048] Also provided here are methods of treating multiple sclerosis (MS) in a subject in need thereof by administering to the subject a therapeutically effective amount of a composition or pharmaceutical formulation described herein, wherein administration of the composition or the pharmaceutical formulation results in no evidence of disease activity (NED A) in the subject 24-96 weeks after the administration.
[0049] In some embodiments, administration of the composition or the pharmaceutical formulation results in NED A in the subject 24 weeks after the administration.
[0050] Also provided here are methods of treating multiple sclerosis (MS) in a subject in need thereof by administering to the subject a therapeutically effective amount of a composition or pharmaceutical formulation described herein, wherein administration of the composition or the pharmaceutical formulation results in a transient decrease in lymphocyte count in the subject. [0051] In some embodiments, the lymphocyte count is normalized by day 8 of the administration.
[0052] In some embodiments, the MS is a relapsing form of MS (RMS).
[0053] Also provided here are methods of reducing annualized relapse rate (ARR) in a subject with relapsing forms of multiple sclerosis (MS) by administering to the subject an effective amount of a composition or pharmaceutical formulation described herein, the method comprising: administering an intravenous infusion of the composition or the pharmaceutical formulation in a multi-infusion dosage regimen, the dosage regimen comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody protein at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody protein at about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the prior infusion.
[0054] In some embodiments, the effective amount of the composition or the pharmaceutical formulation is sufficient to result in an ARR of 0.091 or an ARR of 0.076.
[0055] In some embodiments, duration of the second infusion, the first subsequent infusion, and the one or more subsequent infusions of the anti-CD20 antibody protein is about one hour. [0056] Also provided here are methods of treating relapsing forms of multiple sclerosis (MS) in a subject in need thereof by administering to the subject an effective amount of a composition or pharmaceutical formulation described herein, the method comprising: administering an intravenous infusion of the composition or the pharmaceutical formulation in a multi-infusion dosage regimen, the dosage regimen comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody protein at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody protein at about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the prior infusion, wherein duration of the second infusion, the first subsequent infusion, and the one or more subsequent infusions of the anti-CD20 antibody protein is about one hour.
[0057] In some embodiments, the method further comprises pre-medicating the subject with a corticosteroid and an antihistamine, 30-60 minutes prior to administration of the composition or the pharmaceutical formulation. In some embodiments, the corticosteroid is methylprednisone or dexamethasone. In some embodiments, the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
[0058] In some embodiments, the intravenous infusion of the composition or the pharmaceutical formulation is prepared in 250 mb of 0.9% Sodium Chloride Injection. [0059] In some embodiments, the first subsequent infusion is at about 24 weeks from the first infusion. In some embodiments, the one or more subsequent infusions is at about 24 weeks from the prior infusion. In some embodiments, the first subsequent infusion is at about 6 months from the first infusion. In some embodiments, the one or more subsequent infusions is at about 6 months from the prior infusion.
[0060] In some embodiments, duration of the first infusion of the anti-CD20 antibody protein is about four hours. In some embodiments, the first infusion of the anti-CD20 antibody protein is infused at a rate of 10 mb per hour for the first 30 minutes; 20 mb per hour for the next 30 minutes; 35 mb per hour for the next hour; and 100 mb per hour for the remaining two hours.
[0061] In some embodiments, the second infusion, the first subsequent infusion, and the one or more subsequent infusion of the anti-CD20 antibody protein is infused at a rate of 100 mb per hour for the first 30 minutes, and 400 mb for the remaining 30 minutes.
[0062] In some embodiments, the multi-infusion dosage regimen of the anti-CD20 antibody protein alleviates or delays progression of the symptoms of MS.
[0063] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of gadolinium-enhancing T1 lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0064] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of new and enlarging T2 hyperintense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0065] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased no evidence of disease activity (NED A) status, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0066] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased Confirmed Disability Improvement (CDI), as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0067] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an increased Multiple Sclerosis Functional Composite (MSFC) score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. [0068] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved timed 25-Foot Walk (T25FW) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0069] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved 9-Hole Peg test (9-HPT) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0070] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a significant reduction in both volume and number of new T1 hypointense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[0071] In some embodiments, the multi-infusion dosage regimen of the anti-CD20 antibody protein results in a geometric mean steady-state AUC of 3000 mcg/mL per day (CV = 28%) and a mean maximum concentration of 139 mcg/mL (CV =15%).
[0072] Also provided here are methods for inactivating a virus or adventitious agents in rat myeloma cells expressing the anti-CD20 antibody proteins recited in the composition described herein, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
[0073] Also provided here are methods for reducing immunogenicity of the anti-CD20 antibody proteins recited in the composition described herein, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
[0074] Also provided herein are methods of treating multiple sclerosis, wherein the methods comprise administering a therapeutically effective amount of a pharmaceutical formulation comprising a population of anti-CD20 antibody proteins to a subject in need thereof, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein administration of the pharmaceutical formulation results in no evidence of disease activity (NED A) in the subject 24 weeks after the administration. In some embodiments, administration of the pharmaceutical formulation results in NED A in the subject 24-96 weeks after the administration. [0075] Also disclosed herein are methods of treating multiple sclerosis, wherein the methods comprise administering a therapeutically effective amount of a pharmaceutical formulation comprising a population of anti-CD20 antibody proteins to a subject in need thereof, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein administration of the pharmaceutical formulation results in a transient decrease in lymphocyte count in the subject. In some embodiments, the lymphocyte count is normalized by day 8 of the administration.
[0076] In some embodiments of any of the methods described herein, the population of anti- CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans.
[0077] In some embodiments of any of the methods described herein, the multiple sclerosis (MS) is a relapsing form of MS (RMS). In certain embodiments, the RMS comprises a clinically isolated syndrome ("CIS"); relapsing-remitting MS ("RRMS"); or active secondary progressive MS ("SPMS"). In some embodiments of any of the methods described herein, the subject is diagnosed with RMS in accordance to McDonald Criteria (2010), or by another suitable method known by one skilled in the art. In some embodiments of any of the methods described herein, the subject is a human.
[0078] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans.
[0079] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 35% fucosylated glycans. [0080] In some embodiments, the N-glycan profile comprises 28 to 33% fucosylated glycans. In some embodiments, the N-glycan profile comprises about 30% fucosylated glycans. [0081] In some embodiments, the N-glycan profile comprises 16 to 18% galactosylated glycans. In some embodiments, the N-glycan profile comprises about 17% galactosylated glycans.
[0082] In some embodiments, the relative abundance of fucosylated glycans is the percent of fucosylated glycans among all glycans in the N-glycan profile. In some embodiments, the relative abundance of galactosylated glycans is the percent of galactosylated glycans among all glycans in the N-glycan profile.
[0083] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising at least about 10% bisecting N-glycans.
[0084] In some embodiments, the N-glycan profile comprises 12% to 30% bisecting N- glycans. In some embodiments, the N-glycan profile comprises about 18% bisecting N-glycans. In some embodiments, the bisecting N-glycan comprises one or more of GOB, G0FB, G1FB, G2FBS1, and G2FBS2.
[0085] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising less than 5% sialylated glycans.
[0086] In some embodiments, the N-glycan profile comprises less than 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycan. In some embodiments, the N-glycan profile comprises no detectable amount of sialylated glycan.
[0087] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 5 to 15% GOB N-glycan.
[0088] In some embodiments, the N-glycan profile comprises 9 to 11% GOB N-glycan. In some embodiments, the N-glycan profile comprises about 10% GOB N-glycan.
[0089] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 0.1% to 1.5% Man5 N-glycan.
[0090] In some embodiments, the N-glycan profile comprises 0.4% to 0.7% Man5 N-glycan. In some embodiments, the N-glycan profile comprises about 0.6% Man5 N-glycan. In some embodiments, Man5 N-glycan is the only high mannose species in the N-glycan profile.
[0091] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins comprises 0.20 to 0.40 mol isoaspartate per mol protein.
[0092] In some embodiments, the population of anti-CD20 antibody proteins comprises 0.25 to 0.35 mol isoaspartate per mol protein.
[0093] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the glutamate at position 1 of the heavy chain is a pyroglutamate and wherein the glutamate at position 1 of the light chain is a pyroglutamate.
[0094] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.1 to 0.15 G1 to GO N-glycans.
[0095] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.5 to 0.9 GIF to G1 N-glycans.
[0096] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance range: (a) 0.3% to 2% G0-GN; (b) 0.1% to 2% G0F-GN; (c) 0.1% to 1% Gl-GN; (d) 5% to 20% GOB; (e) 5% to 30% G0F; (f) 0.1% to 1.5% Man5; (g) 1% to 15% G0FB; (h) 1% to 13% Gl; (i) 0.5% to 10% GF; (j) 0.5% to 6% GIB; (k) 0.5% to 12% GIF; (1) 0.1% to 3% GIF’; (m) 0.1% to 3% G1FB; (n) 0.1% to 2% G2; and (o) 0.1% to 2% G2F.
[0097] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance range: (a) 0.8% to 1.1% G0-GN; (b) 0.5% to 1.1% G0F-GN; (c) 0.3% to 0.6% Gl-GN; (d) 9.5% to 14.1% GOB; (e) 12.8% to 19.7% G0F; (f) 0.4% to 0.7% Man5; (g) 5.1% to 7.0% G0FB; (h) 5.7% to 6.4% Gl; (i) 2.7% to 3.3% GF; (j) 1.4% to 2.0% GIB; (k) 2.6% to 4.2% GIF; (1) 1.1% to 1.6% GIF’; (m) 1.1% to 1.8% G1FB; (n) 0.5% to 0.7% G2; and (o) 0.3% to 0.5% G2F.
[0098] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance: (a) 0.9% G0-GN; (b) 0.8% G0F-GN; (c) 0.5% Gl-GN; (d) 10.9% GOB; (e) 17.0% G0F; (f) 0.6% Man5; (g) 6.0% G0FB; (h) 6.1% Gl; (i) 2.9% GF; (j) 1.6% GIB; (k) 3.2% GIF; (1) 1.3% GIF’; (m) 1.3 G1FB; (n) 0.5% G2; and (o) 0.3% G2F.
[0099] In some embodiments, the population of anti-CD20 antibody proteins further comprises at least three, four or five N-glycans within the relative abundance or relative abundance range as described herein.
[00100] In some embodiments, the N-glycan profile of the population of anti-CD20 antibody proteins is determined using a method comprising: (a) incubate the population of anti-CD20 antibody proteins with an enzyme, wherein the enzyme catalyzes releasing of the N-glycans from the anti-CD20 antibody; (b) measure the relevant abundance of the released N-gylcans using one or more methods selected from chromatography, mass spectrometry, capillary electrophoresis, and the combination thereof. In some embodiments, the enzyme is PNGase F.
[00101] In some embodiments, the method further comprises after step (a) and before step (b) the following steps: (c) purify the N-glycans; and (d) label the N-glycans with a fluorescent compound. In some embodiments, the fluorescent compound is 2-aminobenzamide (2-AB).
[00102] In some embodiments, less than 10% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 5% of the anti-CD20 antibody proteins in the population is non-glycosylated. In some embodiments, less than 1% of the anti- CD20 antibody proteins in the population is non-glycosylated.
[00103] In some embodiments, the N-glycan profile of the population of anti-CD20 antibody proteins is substantially as shown in Figure 2.
[00104] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO:1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) 8.0% to 10.0% a-helix; (b) 32.0% to 36.0% Anti-parallel |3-sheet; (c) 5.0% to 6.0% Parallel |3-sheet; (d) 16.0% to 18.0% |3- Turn; and (e) 35.0% to 36.0% random coil.
[00105] In some embodiments, the population of anti-CD20 antibody proteins comprises secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) 8.0% to 10.0% a-helix; (b) 32.0% to 36.0% Anti-parallel |3-sheet; (c) 5.0% to 6.0% Parallel |3- sheet; (d) 16.0% to 18.0% |3-Turn; and (e) 35.0% to 36.0% random coil.
[00106] In some embodiments, the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows: (a) about 9.0% a-helix; (b) about 33.0% Anti-parallel |3-sheet; (c) about 5.6% Parallel |3- sheet; (d) about 17.5% |3-Turn; and (e) about 35.2% random coil.
[00107] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain (“HC”) comprising the amino acid sequence of SEQ ID NO: 1 and a light chain (“LC”) comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins further comprises one or more of the following post-translational modifications at the specified abundance:
Figure imgf000022_0001
[00108] In some embodiments, the population of anti-CD20 antibody proteins comprises two, three, four, five or more of the post translational modifications.
[00109] In some embodiments, the population of anti-CD20 antibody proteins comprises the following post-translational modification at the specified abundance:
Figure imgf000023_0001
[00110] In some embodiments, the population of anti-CD20 antibody proteins comprises the following post-translational modification at the specified abundance:
Figure imgf000023_0002
[00111] In some embodiments, one or more of the post-translational modifications are measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS). In some embodiments, the deamidation is measured by isoaspartate detection or peptide mapping using LC-MS.
[00112] Also provided herein are compositions comprising a population of anti-CD20 antibody proteins described herein, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain (“HC”) comprising the amino acid sequence of SEQ ID NO: 1 and a light chain (“LC”) comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has the following properties:
Figure imgf000023_0003
[00113] In some embodiments, the population has an amount of total protein of 25.5-25.8 mg/mL as measured by absorbance at 280 nm. In some embodiments, the population has an amount of total protein of about 25.6 mg/mL as measured by absorbance at 280 nm. [00114] In some embodiments, the population induces greater cytotoxicity in a cell-based antibody-dependent cellular cytotoxicity (ADCC) assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab. In some embodiments, the population has a relative potency of 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 117% in a cell-based ADCC assay compared to a commercial reference standard. In some embodiments, the cell-based ADCC assay uses effector cells selected from CD 16 effector cells and primary NK cells. In some embodiments, the population performs in a cell-based ADCC using CD 16 effector cells at more than 100% of that of a commercial reference standard.
[00115] In some embodiments, the population exhibits greater B cell depletion activity in a human whole blood B cell depletion assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
[00116] In some embodiments, the population has a relative potency of 78% to 116% or 73% to 128% in a cell-based complement dependent cytotoxicity (CDC) assay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of about 91% in a cell-based CDC assay compared to that of a commercial reference standard. [00117] In some embodiments, the population has a relative potency of 92 to 118% or 82 to 138% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In some embodiments, the population has a relative potency of about 109% in a cellbased CD20 binding activity bioassay compared to that of a commercial reference standard. [00118] In some embodiments, the population has a KD value 30 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In some embodiments, the population has a KD value about 59 nM in an FcyRIIIa- 158 V binding assay as measured by surface plasmon resonance.
[00119] In some embodiments, the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In some embodiments, the population has a KD value 760 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
[00120] In some embodiments, the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab. [00121] In some embodiments, the population has a relative potency of 88 to 113% or 86 to 116% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 99% in a Cl q binding assay as measured by ELISA compared to a commercial reference standard.
[00122] In some embodiments, the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 115% in a CD 16 activity assay compared to a commercial reference standard.
[00123] In some embodiments, the population of anti-CD20 antibody proteins has a purification profile as follows:
Figure imgf000025_0001
[00124] In some embodiments, the population has 99.2 to 99.9% monomers as detected by size exclusion chromatography (SEC). In some embodiments, the population has 0.1 to 0.8% dimers as detected by SEC. In some embodiments, the population has undetectable level of aggregates as detected by SEC. In some embodiments, the population has undetectable level of fragments as detected by SEC.
[00125] In some embodiments, the population has 93.6 to 95.9% IgG after purification by non-reduced capillary gel electrophoresis (CGE). In some embodiments, the population has 0.1 to 0.3% high molecular weight species (HMWS) after purification by non-reduced CGE. In some embodiments, the population has 0.7 to 1.2% free light chain (LC) after purification by non-reduced CGE.
[00126] In some embodiments, the population has 97.7 to 98.0% heavy chain plus light chain species (HC + LC) after purification by reduced CGE. [00127] In some embodiments, the population of anti-CD20 antibody proteins has a distribution of charged isoforms as follows:
Figure imgf000026_0001
[00128] In some embodiments, the population has 20 to 25% acidic isoforms as detected by imaged capillary isoelectric focusing (iCIEF). In some embodiments, the population has 50 to 60% main isoforms as detected by iCIEF. In some embodiments, the population has 20 to 30% basic isoforms as detected by iCIEF.
[00129] In some embodiments, the population has an average molar ratio of free thiol to anti- CD20 antibody of about 2.0 to 2.2.
[00130] In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of the N-terminal residue. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 5 N- terminal residues. In some embodiments, the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of up to 10 N-terminal residues.
[00131] In some embodiments, the terminal lysine amino acid residue of the heavy chain in the anti-CD20 antibody in said population is truncated.
In some embodiments, the administration of the anti-CD20 antibody to a human patient results in one or more of the following pharmacokinetic parameters: (a) an AUC between 2,160 pg/mL and 3,840 pg/mL; (b) a Cmax between 118,011 ng/mL and 159,989 ng/mL; (c) a Cmin between 40 ng/mL and 375 ng/mL; and (d) a Cavg is between 6,437 ng/mL and 11,443 ng/mL, and wherein the anti-CD20 antibody is administered as i) a first infusion at a dose of about 150 mg, ii) a second infusion two weeks later at a dose of about 450 mg, and iii) subsequent infusions every six months at a dose of about 450 mg.
[00132] In some embodiments, the administration of the anti-CD20 antibody to a human patient results in one or more of the following pharmacokinetic parameters: (a) an AUC about 3,000 pg/mL; (b) a Cmax about 139,000 ng/mL; (c) a Cmin about 139 ng/mL; and (d) a Cavg about 8,940 ng/mL. [00133] In some embodiments, the antibody proteins of the population are present in a single dosage form.
[00134] Also provided here are pharmaceutical formulations comprising compositions described herein, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL.
[00135] Also provided here are pharmaceutical formulations comprising anti-CD20 antibody described herein, wherein the anti-CD20 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the pharmaceutical formulation comprises one or more of the following: sodium chloride, trisodium citrate dehydrate, polysorbate 80, and hydrochloric acid.
[00136] In some embodiments, the pharmaceutical formulation comprises about 9.0 mg/mL of sodium chloride. In some embodiments, the pharmaceutical formulation comprises about 7.4 mg/mL of trisodium citrate dehydrate. In some embodiments, the pharmaceutical formulation comprises about 0.7 mg/mL of polysorbate 80. In some embodiments, the pharmaceutical formulation comprises about 0.4 mg/mL of hydrochloric acid.
[00137] Also provided here are single batch preparations of a population of anti-CD20 antibody proteins described herein, wherein the anti-CD20 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the single batch comprises at least 100 g of the anti- CD20 antibody proteins.
[00138] Also provided here are single batch preparations of a population of anti-CD20 antibody proteins described herein, wherein the anti-CD20 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the single batch comprises at least 120 g of the anti- CD20 antibody proteins.
[00139] Also provided here are single batch preparations of a population of anti-CD20 antibody proteins described herein, wherein the anti-CD20 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the single batch comprises at least 150 g of the anti- CD20 antibody proteins. [00140] Also provided here are populations of anti-CD20 antibody proteins described herein, produced in a 15,000 L or 20,000 L bioreactor.
[00141] Also provided here are methods of treating an autoimmune disease, wherein the method comprises administering the compositions described herein to a subject in need thereof, and wherein the autoimmune disease is selected from the group consisting of psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, osteoporosis, eczema, allogenic or xenogenic transplantation, graft rejection, graft- versus-host disease, lupus erythematosus, inflammatory disease, type 1 diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis, myasthenia gravis, autoimmune hemolytic anemia, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic conjunctivitis, atopic dermatitis, chronic obstructive pulmonary disease, glomerulonephritis, neuroinflammatory diseases, and uveitis.
[00142] Also provided here are methods of treating multiple sclerosis, wherein the method comprises administering the compositions described herein to a subject in need thereof.
[00143] In some embodiments, the multiple sclerosis (MS) is a relapsing form of MS. In some embodiments, the relapsing form of MS is selected from clinically isolated syndrome (CIS), relapsing-remitting MS (RRMS) and active secondary progressive MS (SPMS). In some embodiments, the relapsing form of MS is a clinically isolated syndrome (CIS). In some embodiments, the relapsing form of MS is relapse-remitting multiple sclerosis (RRMS). In some embodiments, the relapsing form of MS is active secondary progressive multiple sclerosis (SPMS).
[00144] Also provided here are methods of treating a neoplastic disease, wherein the method comprises administering the compositions described herein to a subject in need thereof.
[00145] In some embodiments, the neoplastic disease is acute B lymphoblastic leukaemia, B- cell lymphoma, mature B-cell lymphoma, including B-type Chronic Lymphocytic Leukaemia (B-CLL), small B-cell lymphoma, B-cell prolymphocytic leukaemia, lymphoplasmocytic lymphoma, mantle cell lymphoma, follicular lymphoma, marginal zone MALT-type lymphoma, lymph node marginal zone lymphoma with or without monocytoid B cells, splenic marginal zone lymphoma (with or without villous lymphocytes), tricholeucocytic leukaemia, diffuse large B- cell lymphoma, or Burkitt's lymphoma. [00146] In some embodiments, the subject is a human.
[00147] Also provided here are methods for inactivating a virus or adventitious agents in rat myeloma cells expressing the anti-CD20 antibody proteins in the compositions described herein, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
[00148] Also provided here are methods of reducing immunogenicity of the anti-CD20 antibody proteins in the compositions described herein, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
[00149] Also provided here are methods of analyzing an TG-1101 (TG Therapeutics, Inc.) preparation, comprising:
(i) providing an isolated N-glycan fraction from the TG-1101 (TG Therapeutics, Inc.) preparation;
(ii) analyzing the N-glycan fraction to determine if one or more N-glycans are the following N- glycans within the following relative abundance range: (a) 0.3% to 2% G0-GN; (b) 0.1% to 2% G0F-GN; (c) 0.1% to 1% Gl-GN; (d) 5% to 20% GOB; (e) 5% to 30% G0F; (f) 0.1% to 1.5% Man5; (g) 1% to 15% G0FB; (h) 1% to 13% Gl; (i) 0.5% to 10% Gl’; (j) 0.5% to 6% GIB; (k) 0.5% to 12% GIF; (1) 0.1% to 3% GIF’; (m) 0.1% to 3% G1FB; (n) 0.1% to 2% G2; and (o) 0.1% to 2% G2F.
In some embodiments, the method comprises analyzing the N-glycan fraction to determine if one or more N-glycans are the following N-glycans within the following relative abundance range:
(a) 0.8% to 1.1% G0-GN; (b) 0.5% to 1.1% G0F-GN; (c) 0.3% to 0.6% Gl-GN; (d) 9.5% to 14.1% GOB; (e) 12.8% to 19.7% G0F; (f) 0.4% to 0.7% Man5; (g) 5.1% to 7.0% G0FB; (h) 5.7% to 6.4% Gl; (i) 2.7% to 3.3% Gl’; (j) 1.4% to 2.0% GIB; (k) 2.6% to 4.2% GIF; (1) 1.1% to 1.6% GIF’; (m) 1.1% to 1.8% G1FB; (n) 0.5% to 0.7% G2; and (o) 0.3% to 0.5% G2F.
[00150] In some embodiments, the method comprises analyzing the N-glycan fraction to determine if one or more N-glycans are in the following relative abundance: (a) 0.9% G0-GN;
(b) 0.8% G0F-GN; (c) 0.5% Gl-GN; (d) 10.9% GOB; (e) 17.0% G0F; (f) 0.6% Man5; (g) 6.0% G0FB; (h) 6.1% Gl; (i) 2.9% Gl’; (j) 1.6% GIB; (k) 3.2% GIF; (1) 1.3% GIF’; (m) 1.3 G1FB; (n) 0.5% G2; and (o) 0.3% G2F.
[00151] Also provided here are compositions comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance: (a) 0.9% GO-GN; (b) 0.8% GOF-GN; (c) 0.5% Gl-GN; (d) 10.9% GOB; (e) 17.0% G0F; (f) 0.6% Man5; (g) 6.0% G0FB; (h) 6.1% Gl; (i) 2.9% G1 ’ ; (j) 1-6% GIB; (k) 3.2% GIF; (1) 1.3% GIF’; (m) 1.3 G1FB; (n) 0.5% G2; and (o) 0.3% G2F, and wherein the population of anti-CD20 antibody proteins is made in a rat hybridoma cell.
[00152] In some embodiments, the rat hybridoma cell is YB2/0 cell.
6. BRIEF DESCRIPTION OF THE DRAWINGS
[00153] Figure 1 illustrates the structure and abbreviations for various N-glycans.
[00154] Figure 2 illustrates the glycosylation profile of a sample of anti-CD20 antibody proteins provided herein.
[00155] Figure 3 illustrates the intact MS Spectra of a sample of anti-CD20 antibody proteins provided herein.
[00156] Figure 4 illustrates antibody-dependent cellular cytotoxicity (ADCC) activity dose response curves using Raji cells and KILR CD16a cells.
[00157] Figure 5 illustrates ADCC activity dose response curves using Raji cells and primary NK cells.
[00158] Figure 6 illustrates antibody-dependent cellular phagocytosis (ADCP) activity dose response curves.
[00159] Figure 7 illustrates complement dependent cytotoxicity (CDC) activity dose response curves.
[00160] Figure 8 illustrates CD20 binding dose response curves.
[00161] Figure 9 illustrates CD20 binding dose response curves using FACS.
[00162] Figure 10 illustrates Clq binding dose response curves.
[00163] Figures 11A-C illustrate human whole blood B cell depletion from three donors. B cell depletion calculated based on data using CD 19 as B cell marker.
[00164] Figure 12 illustrates the Goodness-of-Fit (GOF) diagnostics of the TG-1101 final model. [00165] Figure 13 illustrates the pcVPC for the TG-1101 final PK model by study. The blue dots are prediction-corrected observed concentrations; the blue lines are the 50th (solid), 5th (dashed), and 95th (dashed) percentiles of observed concentrations; and the black lines are the 50th (solid), 5th (dashed), and 95th (dashed) percentiles of simulations. The gray bands are the 95% Pls for the corresponding black lines based on 500 simulations. The short yellow lines indicate bin intervals. The numbers 201,301,302 and 101-304-703 represent the study numbers, TG1101-RMS201, TG1101-RMS301 and TG1101-RMS302 and the studies in the previous analysis set in subjects with hematologic malignancies, respectively. Abbreviations: pcVPC - prediction-corrected visual predictive check; PI - prediction interval; pred-corr - prediction- corrected; popPK - population pharmacokinetic(s).
[00166] Figure 14 illustrates the forest plot of covariate effects on TG-1101 drug exposure. The first and second dashed vertical lines correspond to ratios of 0.8 and 1.25, respectively. The solid vertical line corresponds to a ratio of 1 and represents a typical subject. Points and whiskers represent the estimate and 90% confidence interval, respectively. The blue-gray horizontal bar shows the range of exposures due to between-subject variability. A typical subject is defined as a male subject from North America/Western Europe with a body weight of 73 kg, is ADA negative and has been on treatment for < 416 days. Abbreviations: ADA - antidrug antibody; AUCss - area under the serum TG-1101 concentration-time curve at steady state; BSV - between-subject or inter-individual variability; CI - confidence interval; CMAXSS - maximum TG-1101 concentration at steady state; CMINSS - minimum TG-1101 concentration at steady state; N/A - not applicable.
[00167] Figure 15 depicts the map of expression vector, HK463-25, containing the immunoglobulin heavy and light chain cDNA sequences of source anti-CD20 antibody, TG- 1101, described herein.
[00168] Figures 16A-16C depict the onset and maintenance of no evidence of disease activity (NED A) with ublituximab. Figure 16A is a bar graph showing NEDA-3 rates by treatment EPOCH (Weeks 0-24 vs Weeks 24-96). denominator based on participants in the Weeks 24-96 analysis. Pooled post hoc analysis. Modified intention-to-treat population. Figure 16B is a bar graph showing NEDA-3 rates by treatment EPOCH (Weeks 24-48 vs Weeks 48-96). denominator based on participants in the Weeks 48-96 analysis. Pooled post hoc analysis. Modified intention-to-treat population. Figure 16C is a bar graph showing components driving EDA in ublituximab-treated participants3. 3 Participants may have >1 component of EDA. Pooled post hoc analysis. Modified intention-to-treat population. CDP, confirmed disability progression; EDA, evidence of disease activity; Gd+, gadolinium-enhancing; n/e, new/enlarging; NED A, no evidence of disease activity; NEDA-3, 3-parameter NED A; EPOCH, combination of etoposide phosphate, prednisone, vincristine sulfate (Oncovin), cyclophosphamide, and doxorubicin hydrochloride (hydroxy daunorubicin).
[00169] Figures 17A-17C depict characterization of cytopenias with ublituximab in the ULTMATE I and II Phase 3 studies in participants with relapsing multiple sclerosis. Figure 17A is a bar graph showing hematologic laboratory abnormalities in ublituximab-treated participants (all study visits). a(493/543). Percentages are based on number of participants with non-missing baseline and at least 1 postbaseline assessment. Pooled post hoc analysis. Safety population. Figure 17B is a bar graph showing hematologic laboratory abnormalities in ublituximab-treated participants (excluding Day 2). a(l 75/543). Percentages are based on number of participants with non-missing baseline and at least 1 postbaseline assessment. Pooled post hoc analysis. Safety population. Figure 17C is a bar graph showing proportion of ublituximab-treated participants with low lymphocyte count by visit. a(476/523). Pooled post hoc analysis. Safety population. Percentages are based on number of participants in the population/treatment group. BL, baseline.
[00170] Figures 18A-18B are bar graphs showing the relative reduction in Annualized Relapse Result (ARR) in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The ARR results from ULTIMATE I (Figure 18A) and ULTIMATE II (Figure 18B) are shown.
[00171] Figures 19A-19B are bar graphs showing the relative reduction in Total Number of Gadolinium (Gd)+ T1 Lesions on MRI in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 19A) and ULTIMATE II (Figure 19B) are shown.
[00172] Figures 20A-20B are bar graphs showing the relative reduction in the number of New or Enlarging T2 Lesions on MRI in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 20A) and ULTIMATE II (Figure 20B) are shown.
[00173] Figures 21A-21B are bar graphs showing the improvement in the proportion (%) of patients with No Evidence of Disease Activity (NED A) status in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The NEDA results from ULTIMATE I (Figure 21A) and ULTIMATE II (Figure 21B) are shown.
[00174] Figures 22A-22B are graphs comparing the percentage of RMS patients having Confirmed Disability Progression (CDP) in a pre-specified pooled analysis of patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. Figure 22A shows 12- week CDP and Figure 22B shows 24-week CDP.
[00175] Figures 23A-23B are graphs comparing the percentage of RMS patients achieving Confirmed Disability Improvement (CDI) in a pre-specified pooled analysis of patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. Figure 23A shows 12- week CDI and Figure 23B shows 24-week CDI.
[00176] Figures 24A-24B are bar graphs showing significant improvements from baseline to 96 weeks in mean Multiple Sclerosis Functional Composite (MSFC) scores in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 24A) and ULTIMATE II (Figure 24B) are shown.
[00177] Figures 25A-25B are bar graphs showing significant improvements in mean Timed 25-Foot Walk (T25FW) scores from baseline to 96 weeks in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 25A) and ULTIMATE II (Figure 25B) are shown.
[00178] Figures 26A-26B are bar graphs showing the changes in mean Paced Auditory Serial Additional Test (PAS AT) scores from baseline to 96 weeks in RMS patients administered ublituximab and teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 26A) and ULTIMATE II (Figure 26B) are shown. The change in mean PASAT scores was similar between the treatment groups.
[00179] Figures 27A-27B are bar graphs showing significant increases in mean 9-Hole Peg Test (9-HPT) scores from baseline to 96 weeks in RMS patients administered ublituximab vs. teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 27 A) and ULTIMATE II (Figure 27B) are shown.
[00180] Figures 28A-28B are graphs showing Percent Brain Volume Change (PBVC) on MRI from week 24 to week 96 (post-hoc analysis) of RMS patients administered ublituximab and teriflunomide in the Phase 3 clinical studies, ULTIMATE I and ULTIMATE II, described in the Examples. The results from ULTIMATE I (Figure 28A) and ULTIMATE II (Figure 28B) are shown.
7. DETAILED DESCRIPTION OF THE DISCLOSURE
[00181] Provided herein are populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications. The primary amino acid sequences of such antibodies are provided in Section 7.3. Types of such post-translational modifications and their respective abundance in the anti-CD20 antibody populations are described in Section 7.4. Compositions, including single-batch compositions, of such populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications and ranges of purity are described in Section 7.5(e). Assays for quantifying such post-translational modifications in a population of anti- CD20 antibody proteins are described in Section 7.5. Assays for analyzing populations of anti- CD20 antibody proteins are described in Section 7.5. Assays for demonstrating the biological and clinical significance of such post-translational modifications in a population of anti-CD20 antibody proteins are described in Section 7.7(a). Methods of using such populations of anti- CD20 antibody proteins for the treatment and prevention of medical conditions are described in Section 7.8. Pharmacokinetic and pharmacodynamic properties of the compositions provided herein in human patients are described in Section 7.9. Methods of making such populations of anti-CD20 antibody proteins are described in Section 7.10.
[00182] As used herein, “TG-1101” (TG Therapeutics, Inc.) (also known as ublituximab, UBX, UTX, TG-1101, TGTX-1101, Utuxin™, LFB-R603, TG20, EMAB603) is the source antibody for the anti-CD20 antibody described herein having a unique glycosylation profile that is produced by the disclosed methods.
[00183] The source antibody, TG-1101, is a monoclonal antibody that targets epitopes on CD20, e g., IRAHT (SEQ ID NO: 37), and EPAN (SEQ ID NO: 38). See, Fox, E. et al., Mult.
Scler. 27:420-429 (March 2021); Babiker et al., Expert Opin Investig Drugs 27:407-412 (2018);
Cotchett, KR et al., Multiple Sclerosis and Related Disorders 9: 102787 (2021); Miller et al., Blood /20: Abstract No. 2756 (2012); Deng, C. et. al., J. Clin. Oncol. 31 Abstract No. 8575 (2013). TG-1101 is also described in U.S. Patent Nos. 9,234,045 and 9,873,745.
7.1 Abbreviations and Conventions
[00184] The following abbreviations are used throughout this application.
Figure imgf000035_0001
Figure imgf000036_0001
7.2 Definitions
[00185] As used herein, the term “population of anti-CD20 antibody proteins” refers to a composition of anti-CD20 antibody proteins that is being tested for the abundance of post- translational modifications. The individual anti-CD20 antibody proteins in a population can comprise similar or different post-translational modifications. In some embodiments, a population of anti-CD20 antibody proteins refers to all anti-CD20 antibody proteins that are present in a single dosage form. In some embodiments, a population of anti-CD20 antibody proteins refers to all anti-CD20 antibody proteins that are present in a single batch. In some embodiments, a population of anti-CD20 antibody proteins is an amount sufficient to determine whether the batch of anti-CD20 antibody proteins, when compared to a reference standard, meets or fails a predetermined acceptable range of comparison value or values.
[00186] As used herein, the term “single batch” in the context of anti-CD20 antibody proteins refers to a composition derived from a single production or run from a single bioreactor of a specified volume. For example, the anti-CD20 antibody proteins obtained from a single run of a 15,000L bioreactor can be referred to as a single batch. In certain embodiments, the bioreactor has a capacity of at least 100; 200; 300; 400; 500; 750; 1,000; 2,000; 3,000; 4,000; 5,000; 7,500; 10,000; 15,000; 20,000; or at least 25,000 L.
[00187] As used herein, and unless otherwise specified, the term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1 or 2 standard deviations. In certain embodiments, the term “about” or “approximately” means within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.
7.3 Primary Amino Acid Sequence of the Anti-CD20 Antibody
[00188] In some aspects, the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2. Sequences are provided below in the Sequence Table.
[00189] In some aspects, the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 or an amino acid that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99% identical to SEQ ID NO: 1; and a light chain comprising the amino acid sequence of SEQ ID NO:2 or an amino acid that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least 99% identical to SEQ ID NO:2.
[00190] In some aspects, the anti-CD20 antibody proteins provided herein comprises the VH CDR1, CDR2, and CDR3 region of sequences SEQ ID NOs: 3, 4, and 5, and the VL CDR1, CDR2, and CDR3 region of sequences SEQ ID NOs: 8, 9, and 10.
[00191] In some aspects, the anti-CD20 antibody proteins provided herein comprises the VH of SEQ ID NO: 6 and the VL of SEQ ID NO: 11.
[00192] In some aspect, the nucleic acid sequence encoding the heavy chain of the anti-CD20 antibody proteins provided herein comprises the nucleic acid sequence of SEQ ID NO:35. In some aspect, the nucleic acid sequence encoding the light chain of the anti-CD20 antibody proteins provided herein comprises the nucleic acid sequence of SEQ ID NO: 36.
[00193] In some aspects, the anti-CD20 antibody proteins provided herein bind to the same epitope as TG-1101 (TG Therapeutics, Inc.).
[00194] In some aspects, the anti-CD20 antibody proteins provided herein are chimeric immunoglobulin G1 (IgGl) anti-CD20 monoclonal antibody proteins, each comprised of a tetrameric assembly from two light chains (213 amino acids) and two heavy chains (448 amino acids).
[00195] In some aspects, the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2 and comprise a pyroglutamate at position 1 of light chain and/or heavy chain instead of glutamine, thus resulting in an amino acid sequence of SEQ ID NO: 13 for the heavy chain and/or an amino acid sequence of SEQ ID NO: 14 for the light chain.
[00196] In some aspects, the anti-CD20 antibody proteins provided herein are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2 and comprise a deletion of the C-terminal lysine at the heavy chain thus resulting in the amino acid sequence of SEQ ID NO: 15. [00197] In some embodiments, provided herein is a composition or population of anti-CD20 antibody proteins wherein at least 50%, 60%, 70%, 80%, 90%, 95%, or 98% comprises (i) a pyroglutamate at position 1 (instead of glutamine) of the heavy chain, (ii) a pyroglutamate at position 1 (instead of glutamine) of the light chain, and/or (iii) a deletion of the C-terminal lysine of the heavy chain.
[00198] In some aspects, an anti-CD20 antibody is expressed from one or more nucleic acid sequences encoding a light chain comprising the amino acid sequence of SEQ ID NO: 16.
7.4 Anti-CD20 Antibody Compositions
[00199] The anti-CD20 antibody compositions provided herein can be described by various post-translational modifications and/or by their three-dimensional conformation (see Section 7.5(e)). The respective levels of the various post-translational modifications can be quantified as described in Section 7.5. Without being bound by theory, these structural properties of the anti- CD20 antibody compositions provided herein result in the biological and clinical properties described below in Sections 7.7 and 7.9.
[00200] The anti-CD20 antibody compositions produced in vitro have various post- translational modifications. It is understood that each individual anti-CD20 antibody protein may have its own specific pattern of post-translational modifications. To describe the properties of a population of multiple anti-CD20 antibody proteins, the overall presence of a specific post- translational modification can be quantified. Without being bound by theory, the levels of a specific post-translational modification in a population of anti-CD20 proteins can determine the biological and clinical properties of the composition (such as a dose of a pharmaceutical formulation). Without being bound by theory, the post-translational modifications are rendered by the expression in a rat hybridoma cell (e.g., YB2/0 cell) in cell culture.
[00201] In some embodiments, a type of post-translational modification that can be used to describe the anti-CD20 antibody compositions provided herein is glycosylation. Various glycosylations are known. In one aspect, the glycosylation is N-glycosylation. N-glycans that can be present can be any one of the N-glycans shown in Figure 1. Levels of N-glycosylation are discussed in the Section 7.4(a) below and can be quantified using the assays in Section 7.5.
[00202] In some embodiments, a type of post-translational modification that can be used to describe the anti-CD20 antibody compositions provided herein is deamidation. Deamidation is a
31 chemical reaction in which an amide functional group in the side chain of the amino acid asparagine or glutamine is removed or converted to another functional group. Typically, asparagine is converted to aspartic acid or isoaspartic acid. Levels of deamidation at a specific amino acid position in the anti-CD20 antibody compositions provided herein are described in Section 7.4(b) below and can be determined as described in Section 7.5 herein.
(a) N-glycosylation
[00203] Various forms of N-glycosylation can be present in the anti-CD20 antibody compositions provided herein. Without being bound by theory, the relative distribution of the various N-glycans, or individual sugar residues present in those N-glycans, among the individual anti-CD20 antibody proteins in a population of anti-CD20 antibody proteins can determine the biological and clinical properties (such as the biological and clinical properties discussed in Sections 7.7 and 7.9) of the anti-CD20 antibody composition provided herein. The anti-CD20 antibody composition provided herein can be described by any one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty or all of the N-glycans or individual sugar residues described in the following subsections.
[00204] In some embodiments, an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
(a) 0.3% to 2% G0-GN;
(b) 0.1% to 2% G0F-GN;
(c) 30% to 60% GO;
(d) 0.1% to 1% G1-GN;
(e) 5% to 20% GOB;
(f) 5% to 30% G0F;
(g) 0.1% to 1.5% Man5;
(h) l% to 15% G0FB;
(i) l% to 13% Gl;
(j) 0.5% to 10% GL;
(k) 0.5% to 6% GIB;
(l) 0.5% to 12% GIF; (m) 0.1% to 3% GIF’;
(n) 0.1% to 3% G1FB;
(o) 0.1% to 2% G2; and
(p) 0.1% to 2% G2F.
[00205] In some embodiments, an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
(a) 0.8% to 1.1% GO-GN;
(b) 0.5% to 1.1% G0F-GN;
(c) 42.5% to 48.8% GO;
(d) 0.3% to 0.6% Gl-GN;
(e) 9.5% to 14.1% GOB;
(f) 12.8% to 19.7% G0F;
(g) 0.4% to 0.7% Man5;
(h) 5.1% to 7.0% G0FB;
(i) 5.7% to 6.4% Gl;
(j) 2.7% to 3.3% Gl’;
(k) 1.4% to 2.0% GIB;
(l) 2.6% to 4.2% GIF;
(m) 1.1% to 1.6% GIF’;
(n) 1.1% to 1.8% G1FB;
(o) 0.5% to 0.7% G2; and
(p) 0.3% to 0.5% G2F.
[00206] In some embodiments, an anti-CD20 antibody composition provided herein comprises at least two, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, or 15 N- glycans within the following relative abundance range:
(a) 0.9% G0-GN;
(b) 0.8% G0F-GN;
(c) 46.1% GO;
(d) 0.5% Gl-GN;
(e) 10.9% GOB; (f) 17.0% GOF;
(g) 0.6% Man5;
(h) 6.0% GOFB;
(i) 6.1% Gl;
(j) 2.9% Gl’;
(k) 1.6% GIB;
(l) 3.2% GIF;
(m) 1.3% GIF’;
(n) 1.3 G1FB;
(o) 0.5% G2; and
(p) 0.3% G2F.
[00207] In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.3% to about 2% G0-GN, from about 0.8% to about 1.1% G0-GN, or about 0.9% G0-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 2% G0F-GN, from about 0.5% to about 1.1% G0F-GN, or about 0.8% G0F-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 30% to 60% GO, from about 42.5% to 48.8% GO, or about 46.1% GO. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 1% Gl-GN, from about 0.3% to about 0.6% Gl-GN, or about 0.5% Gl-GN. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 5% to about 20% GOB, from about 5% to about 15% GOB, from about 9.5% to about 14.1% GOB, about 10.9% GOB, or about 10% GOB. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 5% to about 30% GOF, from about 12.8% to about 19.7% GOF, or about 17.0% GOF. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 1.5% Man5, from about 0.4% to about 0.7% Man5, or about 0.6% Man5. In some embodiments, Man5 is the only high mannose N-glycan in the N-glycan profile. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 1% to about 15% GOFB, from about 5.1% to about 7.0% GOFB, or about 6.0% GOFB. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 1% to about 13% Gl, from about 5.7% to about 6.4% Gl, or about 6.1% Gl. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 10% Gl ’, from about 2.7% to about 3.3% Gl’, or about 2.9% Gl ’. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 6% GIB, from about 1.4% to about 2.0% GIB, or about 1.6% GIB. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.5% to about 12% GIF, from about 2.6% to about 4.2% GIF, or about 3.2% GIF. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 3% GIF’, from about 1.1% to about 1.6% GIF’, or about 1.3% GIF’. In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 3% G1FB, from about 1.1% to about 1.8% G1FB, or about 1.3 G1FB. In certain embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.1% to about 2% G2, from about 0.5% to about 0.7% G2, or about 0.5% G2. In some embodiments, the population of anti-CD20 antibody proteins comprises a relative abundance of from an N-glycan profile comprising about 0.1% to about 2% G2F, from about 0.3% to about 0.5% G2F, or about 0.3% G2F.
[00208] In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.3% to about 2% G0-GN, from about 0.1% to about 2% G0F-GN, from about 30% to 60% GO, from about 0.1% to about 1% Gl-GN, from about 5% to about 20% GOB, from about 5% to about 30% G0F, from about 0.1% to about 1.5% Man5, from about 1% to about 15% GOFB, from about 1% to about 13% Gl, from about 0.5% to about 10% Gl’, from about 0.5% to about 6% GIB, from about 0.5% to about 12% GIF, from about 0.1% to about 3% GIF’, from about 0.1% to about 3% G1FB, from about 0.1% to about 2% G2, and from about 0.1% to about 2% G2F. In some embodiments, Man5 is the only high mannose N-glycan in the N-glycan profile. [00209] In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of from about 0.8% to about 1.1% GO-GN, from about 0.5% to about 1.1% G0F-GN, from about 42.5% to 48.8% GO, from about 0.3% to about 0.6% Gl-GN, from about 9.5% to about 14.1% GOB, from about 12.8% to about 19.7% G0F, from about 0.4% to about 0.7% Man5, from about 5.1% to about 7.0% G0FB, from about 5.7% to about 6.4% Gl, from about 2.7% to about 3.3% Gl’, from about 1.4% to about 2.0% GIB, from about 2.6% to about 4.2% GIF, from about 1.1% to about 1.6% GIF’, from about 1.1% to about 1.8% G1FB, from about 0.5% to about 0.7% G2, and from about 0.3% to about 0.5% G2F. In some embodiments, Man5 is the only high mannose N-glycan in the N-glycan profile.
[00210] In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance of about 0.9% GO-GN, about 0.8% G0F-GN, about 46.1% GO, about 0.5% Gl-GN, about 10.9% GOB, about 17.0% G0F, about 0.6% Man5, about 6.0% G0FB, about 6.1% Gl, about 2.9% Gl ’, about 1.6% GIB, about 3.2% GIF, about 1.3% GIF’, about 1.3 G1FB, about 0.5% G2, and about 0.3% G2F. In some embodiments, Man5 is the only high mannose N-glycan in the N-glycan profile.
[00211] In some embodiments, the population of anti-CD20 antibody proteins comprises an N-glycan profile comprising a relative abundance ratio of about 0.1 to about 0.15 Gl to GO N- glycans. In some embodiments, the population of anti-CD20 antibody proteins comprises an N- glycan profile comprising a relative abundance ratio of about 0.5 to about 0.9 GIF to Gl N- glycans.
[00212] In one embodiment, an anti-CD20 antibody composition has an N-glycan profile as shown in Figure 2.
[00213] Specific ranges and values for various N-glycans are provided below. The compositions or populations of anti-CD20 antibody proteins provided herein can be described by any one of the prevalence of any of these N-glycans or by any group or by all of these N-glycans.
(i) Galactosylation
[00214] In some embodiments, an anti-CD20 antibody composition provided herein comprises between 10-20% galactosylated glycans. Galactosylated glycans are those N-glycans shown in Figure 1 that carry a galactose residue (shown as open circle in Figure 1). Assays for determining the percentage of galactosylation (or galactosylated N-glycans) are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N- glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of galactosylated N-glycans is the percent of galactosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
[00215] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, or at least 18% and at most 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or at most 20% galactosylated glycans. In some embodiments, an anti-CD20 antibody composition provided herein comprises between 11% and 19%, 12% and 18%, 13% and 17%, or 14% and 16% of galactosylated glycans. For example, an anti-CD20 antibody composition provided herein can comprise about 17% galactosylated glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
(ii) Fucosylation
[00216] In some embodiments, an anti-CD20 antibody composition provided herein comprises between 20% to 40% fucosylated glycans; between 23% to 36% fucosylated glycans; between 20% and 35% fucosylated glycans; between 28% and 33% fucosylated glycans; or about 33% fucosylated glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%). Fucosylated glycans are those N-glycans shown in Figure 1 that carry a fucose residue (shown as open triangle in Figure 1). Assays for determining the percentage of fucosylation (or fucosylated N-glycans) are described in Section 7.5. In short, a sample or a population of anti- CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of fucosylated N-glycans is the percent of fucosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
[00217] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, or at least 33% and at most 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 39%, 40%, 41%, or at most 42% fucosylated glycans.
(iii) Galactosylation to Fucosylation Ratio
[00218] In some embodiments, an anti-CD20 antibody composition provided is characterized by a specified ratio of total fucosylated glycans to total galactosylated glycans (or “Fucose/Galactose ratio”). This Fucose/Galactose ratio can be between 1.5 and 2.1 ; between 1.5 and 2; between 1.5 and 1.9; between 1.5 and 1.8; between 1.6 and 2.1; between 1.7 and 2.1; between 1.8 and 2.8; between 1.6 and 2.0; between 1.7 and 1.9; between 1.6 and 1.8; or the Fucose/Galactose ratio can be about 1.75 (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%). Fucosy lated glycans are those N-glycans shown in Figure 1 that carry a fucose residue (shown as open triangle in Figure 1). Galactosylated glycans are those N-glycans shown in Figure 1 that carry a galactose residue (shown as open circle in Figure 1). Assays for determining the percentage of fucosylation (or fucosylated N-glycans) are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of fucosylated N-glycans is the percent of fucosylated N-glycans among N-glycans that were cleaved using the enzymatic digest.
(iv) Bisecting N-glycans
[00219] In some embodiments, an anti-CD20 antibody composition provided herein comprises at least 10%, 15%, 20%, 25%, or at least 30% bisecting N-glycans; between 10% and 30%, between 12% and 30%, between 12% and 25%, between 12% and 20%, between 15% and 30%, between 15% and 25%, between 15% and 20%, between 18% and 30%, or between 18% and 25% bisecting N-glycans. Bisecting N-glycans are those N-glycans shown in Figure 1 that have a third GlcNAc attached to the mannose residue closest to the protein backbone (shown as open triangle in Figure 1). Assays for determining the percentage of bisecting N-glycans) are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of fucosylated N-glycans is the percent of bisecting N-glycans among N-glycans that were cleaved using the enzymatic digest.
(v) Sialylation
[00220] In some embodiments, an anti-CD20 antibody composition provided herein comprises less than 10%, 8%, 5%, 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycans. In some embodiments, an anti-CD20 antibody composition provided herein comprises between 10% and 0.5% sialylated glycans; between 10% and 5% sialylated glycans; between 5% and 0.5% sialylated glycans; between 4% and 0.5% sialylated glycans; between 2% and 0.5% sialylated glycans; or no detectable amount of sialylated glycans. Assays for determining the percentage of sialylation (or sialylated N-glycans) are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of sialylated N-glycans is the percent of sialylated N- glycans among N-glycans that were cleaved using the enzymatic digest.
[00221] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least no detectable amount, 0.5%, 1%, 2%, 3%, 4%, or at least 5% and at most 0.5%, 1%, 2%, 3%, 4%, 5% or at most 10% sialylated glycans. In some embodiments, an anti-CD20 antibody composition provided herein comprises no detectable amount of sialylated glycans.
(vi) GOB N-glycan
[00222] In some embodiments, an anti-CD20 antibody composition provided herein comprises between 5% and 15% GOB N-glycans; between 9% and 11% GOB N-glycan; or about 10% GOB N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%). GOB N- glycans are shown in Figure 1. Assays for determining the percentage of GOB N-glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of GOB N-glycans is the percent of GOB N-glycans among N-glycans that were cleaved using the enzymatic digest.
[00223] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least 5%, 6%, 7%, 8%, 9%, 10%, or at least 11% and at most 7%, 8%, 9%, 10%, 11%, 12% or at most 13% GOB N-glycans.
(vii) Man5 N-glycan
[00224] In some embodiments, an anti-CD20 antibody composition provided herein comprises between 0.1% and 1.5% Man5 N-glycans; between 0.4% and 0.7% GOB N-glycan; or about 0.6% Man5 N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%). Man5 N-glycans are shown in Figure 1. Assays for determining the percentage of Man5 N-glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of Man5 N-glycans is the percent of Man5 N-glycans among N-glycans that were cleaved using the enzymatic digest.
[00225] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, or at least 0.7% and at most 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, or at most 0.9% Man5 N-glycans.
[00226] In some embodiments, Man5 N-gylcan is the only high mannose species in the N- glycan profile.
(viii) GO N-glycan
[00227] In some embodiments, an anti-CD20 antibody composition provided herein comprises between about 42% and about 52.8% GO N-glycans (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%). GO N-glycans are shown in Figure 1. Assays for determining the percentage of GO N-glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of GO N-glycans is the percent of GO N-glycans among N-glycans that were cleaved using the enzymatic digest.
[00228] In some embodiments, an anti-CD20 antibody composition provided herein comprises between at least 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47% or at least 48% and at most 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 52.8%, 53%, 54%, 55%, 56%, 57% or at most 58% GO N-glycans.
(ix) G1 to GO N-glycans Ratio
[00229] In some embodiments, an anti-CD20 antibody composition provided is characterized by a specified abundance ratio of G1 to GO N-glycans. The abundance ratio of G1 to GO N- glycans can be between 0.02 and 0.3; between 0.05 and 0.25; between 0.08 and 0.22; between 0.09 and 0.2; between 0.1 and 0.19; between 0.1 and 0.18; between 0.1 and 0.17; between 0.1 and 0.16; or the abundance ratio of G1 to GO N-glycans can be between 0.1 and 0.15. G1 and G1 N-glycans are shown in Figure 1. Assays for determining the percentage of G1 or GO glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of G1 or GO glycans is the percent of G1 or GO glycans, respectively, among N-glycans that were cleaved using the enzymatic digest.
(x) GIF to G1 N-glycans Ratio
[00230] In some embodiments, an anti-CD20 antibody composition provided is characterized by a specified abundance ratio of GIF to G1 N-glycans. The abundance ratio of GIF to G1 N- glycans can be between 0.1 and 1.2; between 0.2 and 1.1; between 0.3 and 1; between 0.4 and 1; between 0.5 and 1; or the abundance ratio of G1 to GO N-glycans can be between 0.5 and 0.9. GIF and G1 N-glycans are shown in Figure 1. Assays for determining the percentage of GIF or G1 glycans are described in Section 7.5. In short, a sample or a population of anti-CD20 antibody proteins is subjected to enzymatic deglycosylation so that all N-glycans are cleaved from the core. The resulting N-glycans can subsequently be analyzed, e.g., by mass spectrometry. The percent of GIF or G1 glycans is the percent of GIF or G1 glycans, respectively, among N-glycans that were cleaved using the enzymatic digest.
(b) Deamidation
[00231] In some embodiments, the population of anti-CD20 antibody proteins as described herein comprises asparagine deamidation at one or more asparagine residues present in a heavy chain. In some embodiments, the one or more deamidated asparagine residues present in the heavy chain are selected from Asn-33 (as shown in SEQ ID NO: 18), Asn-55 (as shown in SEQ ID NO:19), Asn-61 (as shown in SEQ ID NO: 19), Asn-160 (as shown in SEQ ID NO:22), Asn-202 (as shown in SEQ ID NO:22), Asn-204 (as shown in SEQ ID NO:22), Asn-277 (as shown in SEQ ID NO:25), Asn-287 (as shown in SEQ ID NO:25), Asn-362 (as shown in SEQ ID NO:27), or Asn-385 (as shown in SEQ ID NO:28). In some embodiments, the population of anti-CD20 antibody proteins as described herein comprises asparagine deamidation at one or more asparagine residues present in a light chain. In some embodiments, the one or more deamidated asparagine residues present in the light chain are selected from Asn-136 (as shown in SEQ ID NO:31), Asn-137 (as shown in SEQ ID NO:31), Asn-151 (as shown in SEQ ID NO:32), or Asn-157 (as shown in SEQ ID NO:32). (c) Oxidation
[00232] In some embodiments, the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a heavy chain. In some embodiments, the one or more methionine residues present in the heavy chain are selected from Met-20, Met-34, Met-81, Met-253 or Met-428 as shown in SEQ ID NO: 17, 18, 21, 24, or 29, respectively. In some embodiments, the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a light chain. In some embodiments, the one or more methionine residues present in the light chain are selected from Met-21 or Met-32 as shown in SEQ ID NO:30.
(d) Pyroglutamation
[00233] In some embodiments, the population of anti-CD20 antibody proteins comprises pyroglutamation at the N-terminal glutamine residue present in a heavy chain or a light chain. In some embodiments, the pyroglutamation at the N-terminal glutamine residue is present in the heavy chain, for example, pGlu-1 as shown in SEQ ID NO: 13. In some embodiments, the pyroglutamation at the N-terminal glutamine residue is present in the light chain, for example, pGlu-1 as shown in SEQ ID NO: 14. In some embodiments, the glutamate at position 1 of the heavy chain is a pyroglutamate and the glutamate at position 1 of the light chain is a pyroglutamate.
(e) Lysine truncation
[00234] In some embodiments, the population of anti-CD20 antibody proteins comprises a deletion of the C-terminal lysine amino acid residue present in a heavy chain or a light chain. In some embodiments, the C-terminal lysine amino acid residue of the heavy chain in the anti- CD20 antibody in said population is truncated. In some embodiments, the population of anti- CD20 antibody proteins comprises a deletion of the C-terminal lysine at the heavy chain. In some embodiments, the heavy chain of the anti-CD20 antibody proteins comprises the amino acid sequence of SEQ ID NO: 15.
(f) Conformation
[00235] Three-dimensional conformation or protein folding can be determined using the assay described in Section 7.5(e). [00236] In some embodiments, a composition or population of anti-CD20 antibody proteins provided herein (e.g., such that are expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2) have a specified three-dimensional folding pattern or confirmation. In some embodiments, the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at a range of 3.0% to 15.0%; Anti-parallel P-sheet at a range of 25.0% to 40.0%; Parallel P-sheet at a range of 0.5% to 12.0%; P-Turn at a range of 10.0% to 25.0%; and random coil at a range of 30.0% to 42.0%.
[00237] In some embodiments, the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at a range of 8.0% to 10.0%; Anti-parallel P-sheet at a range of 32.0% to 36.0%; Parallel P-sheet at a range of 5.0% to 6.0%; P-Turn at a range of 16.0% to 18.0%; and random coil at a range of 35.0% to 36.0%.
[00238] In some embodiments, the population of anti-CD20 antibody proteins comprises two or more of the following secondary structures as determined by circular dichroism (CD) spectroscopy at 205 nm to 260 nm: a-helix at about 9.0%; Anti-parallel P-sheet at about 33.0%; Parallel P-sheet at about 5.6%; P-Turn at about 17.5%; and random coil at about 35.2%, wherein the term “about” means ± 5%.
7.5 Assays for Quantifying Post-Translational Modifications
[00239] In some embodiments, provided herein is a method of determining the amount of post-translational modifications in a population of anti-CD20 antibody proteins. In some embodiments, the post-translational modification is selected from asparagine deamidation, methionine oxidation, glycosylation, pyroglutamate formation, and lysine truncation.
(a) General digestion
[00240] In some embodiments, the method of determining the amount of post-translational modifications in a population of anti-CD20 antibody proteins comprises a step of digesting the population of anti-CD20 antibody proteins with an endoproteinase. In some embodiments, the population of anti-CD20 antibody proteins is reduced prior to digestion. In some embodiments, the population of anti-CD20 antibody proteins is alkylated prior to digestion. In some embodiments, the endoproteinase is selected from Asp-N, Lys-C, or trypsin. In some embodiments, the step of digestion occurs at 37°C for at least 8 hours, at least 12 hours, at least 16 hours, at least 20 hours, overnight, and/or less than 24 hours. In some embodiments, the population of anti-CD20 antibody proteins is digested using Asp-N or Lys-C at 37°C overnight. In some embodiments, the population of anti-CD20 antibody proteins is digested using trypsin at ratio of 50: 1 (w:w) at 37°C overnight. In some embodiments, the digested population of anti- CD20 antibody proteins is purified from digestion reaction components and/or undigested anti- CD20 antibody proteins.
(b) Methods of Determining N-glycosylation
[00241] Exemplary assays and their results are discussed in the Examples in Sections 8.1 and 8.2 below.
[00242] In some embodiments, the method comprises a step for deglycosylating a population of anti-CD20 antibody proteins, thereby producing released N-glycans for labeling. In some embodiments, deglycosylating comprises breaking the glycosidic bond between one or more or all N-glycans from a population of anti-CD20 antibody proteins. In some embodiments, deglycosylating comprises releasing some or most or substantially all of the N-glycans from a population of anti-CD20 antibody proteins. In some embodiments, deglycosylating releases greater than 50 percent, greater than 60 percent, greater than 70 percent, greater than 80 percent, greater than 90 percent, greater than 95 percent, greater than 97 percent, greater than 98 percent, greater than 99 percent, or 100 percent of N-glycans present on a population of anti-CD20 antibody proteins.
[00243] In some embodiments, deglycosylating comprises contacting the population of anti- CD20 antibody proteins with one or more deglycosylating reagent, which cleaves N-glycans or N-linked oligosaccharides. In some embodiments, the deglycosylating reagent is a deglycosylating enzyme or chemical agent. In some embodiments, the deglycosylating enzyme is PNGase F.
[00244] In some embodiments, deglycosylating comprises contacting the population of anti- CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent at a deglycosylation temperature of from about 25°C to about 50°C, from about 37°C to about 50°C, about 25 C, about 37°C, about 42°C, or about 50°C. In some embodiments, the deglycosylation temperature is 37°C. The rate of deglycosylation may be increased by increasing the deglycosylation temperature. In some embodiments, deglycosylation comprises contacting a population of anti-CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent for a period of time sufficient to release some or most or substantially all of the N-glycans from the population of anti-CD20 antibody proteins. In some embodiments, deglycosylating comprises contacting a population of anti-CD20 antibody proteins with said one or more deglycosylating enzyme or chemical agent for at least 30 minutes, at least 1 hour, at least 2 hours, at least 4 hours, at least 8 hours, at least 12 hours, at least 16 hours, at least 20 hours, at least 24 hours, at least 30 hours, and/or less than 48 hours. In some embodiments, deglycosylating comprises contacting a population of anti-CD20 antibody proteins with said one or more deglycosylating enzyme or chemical agent for a period of time of from about 30 minutes to about 2 hours, about 2 hours to about 4 hours, about 4 hours to about 8 hours, about 8 hours to about 12 hours, about 12 hours to about 20 hours, about 20 to about 30 hours, greater than about 30 hours, and/or less than 48 hours. In some embodiments, deglycosylating comprises contacting a population of anti-CD20 antibody proteins with one or more deglycosylating enzyme or chemical agent for about 12 to about 20 hours. In some embodiments, deglycosylation comprises contacting a population of anti-CD20 antibody proteins with PNGase F at about 37 °C for about 12 to about 20 hours. In some embodiments, deglycosylation comprises contacting an anti- CD20 antibody with PNGase F at 37 °C for a time period of from 12-20 hours. In some embodiments, deglycosylation with PNGase F occurs in the presence of a non-ionic detergent (i.e., NP-40).
[00245] In some embodiments, deglycosylation comprises denaturing the population of anti- CD20 antibody proteins prior to contacting the population with said one or more deglycosylating enzyme or chemical agent. In some embodiments, denaturing comprises thermal denaturation, chemical denaturation, or a combination of both. In some embodiments, thermal denaturation comprises incubating a population of anti-CD20 antibody proteins at a denaturation temperature and denaturation time sufficient to unfold some or most or all of the immunoglobulin fold domains of anti-CD20 antibody proteins of said population. In some embodiments, the denaturation temperature is at least 50°C, at least 60 °C, at least 65 °C, at least 70 °C, at least 75 °C, at least 80 °C, at least 85 °C, at least 90 °C, and/or less than about 100 °C. In some embodiments, the denaturation temperature is from about 50 °C to about 60 °C, from about 60 °C to about 70 °C, from about 70 °C to about 80 °C, from about 80 °C to about 90°C, or from 90 °C to about 100 °C. In some embodiments, the denaturation temperature is about 50 °C, about 60 °C, about 65 °C, about 70 °C, about 75 °C, about 80 °C, about 85 °C, about 90 °C, or about 100 °C. In some embodiments, the denaturation temperature is 70 °C. In some embodiments, chemical denaturation comprises incubating a population of anti-CD20 antibody proteins at a denaturation temperature of at least 25 °C, at least 30 °C, at least 37 °C, or an elevated temperature (i.e., a thermal denaturation temperature). In some embodiments, the denaturation time is at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 4 hours, at least 10 hours, or at least 24 hours. In some embodiments, the denaturation time is about 5 minutes, about 10 minutes, about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, about 4 hours, about 10 hours, or about 24 hours. In some embodiments, the denaturation time is 10 minutes. In some embodiments, chemical denaturation comprises contacting the population of anti-CD20 antibody proteins with one or more chemical denaturant. In some embodiments, the one or more chemical denaturant is selected from an ionic detergent, non-ionic detergent, zwitterionic detergent, chaotropic detergent, or reducing agent. In some embodiments, the chemical denaturant is selected from sodium dodecyl sulfate (SDS), urea, or dithiothreitol (DDT). The effects of certain chemical denaturants may interfere with deglycosylation (i.e., enzymatic deglycosylation) and can be countered in solution. In some embodiments, the chemical denaturant is countered by addition of a non-ionic detergent to the solution. In some embodiments, the chemical denaturant is SDS and the additional non-ionic detergent is NP-40. In some embodiments, denaturing a population of anti-CD20 antibody proteins comprises thermal denaturation at 70 °C for 10 minutes.
[00246] In some embodiments, the method comprises purifying released N-glycans of a population of anti-CD20 antibody proteins from a deglycosylation reaction mixture. In some embodiments, the released N-glycans are substantially pure of deglycosylating reagent and deglycosylated or unreacted anti-CD20 antibodies. In some embodiments, the released N- glycans are substantially pure of salts and/or detergents. In some embodiments, the released N- glycans are purified by hydrophilic interaction. In some embodiments, the released N-glycans are purified by chromatography comprising a hydrophilic stationary phase and reversed-phase eluent. In some embodiments, the released N-glycans are purified by hydrophilic interaction liquid chromatography (HILIC). In some embodiments, the released N-glycans are purified via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol. In some embodiments, the released N-glycans are purified to a purity level equivalent to the purity level obtained when purifying via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol.
[00247] In some embodiments, the method comprises a step for labeling released N-glycans of a population of anti-CD20 antibody proteins, thereby producing labeled N-glycans for detection. In some embodiments, labeling released N-glycans comprises chemical derivatization, for example to provide a detectable charge, ultraviolet activity, or fluorescent characteristic to the released N-glycans. In some embodiments, labeling released N-glycans comprises reductive amination, hydrazide labeling, methylation, Michael addition, or permethylation. In some embodiments, labeling released N-glycans comprises contacting the released N-glycans with a label selected from 2-aminobenzamide (2- AB), 2-aminobenzoic acid (2-AA), 2-aminopyridine (PA), 2-aminonaphthalene trisulfonic acid (ANTS), or 1 -aminopyrene- 3,6,8-trisulfonic acid (APTS). In some embodiments, the label is 2-AB. In some embodiments, labeling of released N-glycans by reductive amination comprises the use of a reducing agent. In some embodiments, the reducing agent is selected from sodium cyanoborohydride or 2-picoline borane. In some embodiments, the reducing agent is cyanoborohydride. In some embodiments, labeling released N-glycans by reductive amination comprises contacting the released N-glycans with a label suitable for reductive amination and a reducing agent, at a reaction temperature and reaction time sufficient for labeling to occur. In some embodiments, the reaction temperature is of from about 25 °C to about 40 °C, from about 40 °C to about 50 °C, from about 50 °C to about 60 °C, from about 60 °C to about 70 °C, or from about 70°C to about 80 °C. In certain embodiments, the reaction temperature is about 25 °C, about 30 °C, about 35 °C, about 40 °C, about 45 °C, about 50 °C, about 55 °C, about 60 °C, about 65 °C, about 70 °C, or about 75°C. In some embodiments, the reaction temperature is 65 °C. In some embodiments, the reaction time is at least 30 minutes, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, or at least 6 hours. In some embodiments, the reaction time is of from about 30 minutes to about 1 hour, from about 1 hours to about 2 hours, from about 2 hours to about 3 hours, from about 3 hours to about 4 hours, or from about 4 hours to about 6 hours. In some embodiments, the reaction time is about 30 minutes, about 1 hours, about 2 hours, about 3 hours, about 4 hours, or about 6 hours. In some embodiments, the reaction time is 3 hours. In some embodiments, labeling released N- glycans from a population of anti-CD20 antibody proteins comprises reductive amination using 2- AB and cyanoborohydride at 65 °C for 3 hours, thereby producing labeled N-glycans.
[00248] In some embodiments, the method comprises purifying labeled N-glycans of a population of anti-CD20 antibody proteins from a labeling reaction mixture. In certain embodiments, the labeled N-glycans are substantially pure of unreacted label. In certain embodiments, the labeled N-glycans are substantially pure of unreacted released N-glycans. In certain embodiments, the labeled N-glycans are substantially pure of reducing agent (i.e., cyanoborohydride). In certain embodiments, the labeled N-glycans are purified by hydrophilic interaction. In certain embodiments, the labeled N-glycans are purified by chromatography comprising a hydrophilic stationary phase and reversed-phase eluent. In certain embodiments, the labeled N-glycans are purified by hydrophilic interaction liquid chromatography (HILIC). In some embodiments, the labeled N-glycans are purified via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol. In some embodiments, the labeled N-glycans are purified to a purity level equivalent to the purity level obtained when purifying via a Waters HILIC MassPrep pElution plate, in accordance to manufacturer’s protocol.
[00249] In some embodiments, the method comprises a step for separating labeled N-glycans of a population of anti-CD20 antibody proteins and determining an N-glycan profile of the population. In some embodiments, separating labeled N-glycans comprises separation by hydrophilicity. In some embodiments, separating labeled N-glycans comprises chromatography, which comprises a hydrophilic stationary phase and a reversed-phase eluent. In specific embodiments, the chromatography is hydrophilic interaction liquid chromatography (HILIC). In some embodiments of the method, separating labeled N-glycans comprises chromatography comprising an amide stationary phase. In some embodiments, the amide stationary phase is a glycan BEH amide column. In certain embodiments, the reversed-phase eluent comprises one or more mobile phase, wherein the mobile phase comprises an acidic ammonium buffer and/or acetonitrile.
[00250] In some embodiments, chromatography is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate labeled N-glycans. In some embodiments, chromatography is performed using a chromatography system that controls these parameters (i.e., an HPLC or UPLC system). In some embodiments, the chromatography system is coupled to a detector, for detecting labeled N-glycans of a population of anti-CD20 antibody proteins. In some embodiments, the detector is a fluorescence detector that detects fluorescence from the label (e.g., the label that is contacted with the released N-glycans of a population of anti-CD20 antibody proteins in a labeling step). In some embodiments, the label is 2-AB. In some embodiments, detecting fluorescence comprises exciting with 360 nm wavelength light and observing fluorescent emission at 428 nm wavelength light. In some embodiments, the chromatography system is a HILIC-UPLC system. In some embodiments, separation and detection of labeled N-glycans is performed using a hydrophilic interaction stationary phase, a revers ed-phase eluent, and a Waters UPLC equipped with fluorescence detector. In some embodiments, the hydrophilic interaction stationary phase comprises a glycan BEH amide column (130A, 1.7 pm, 2.1 mm X 150 mm). In some embodiments, the stationary phase (i.e., column) temperature is 50 °C. In some embodiments, the flow rate is 0.50 mL/min. In some embodiments, the reversed-phase eluent comprises one or more mobile phases comprising a first mobile phase comprising about 250 mM ammonium formate, at a pH of about 4.4, and a second mobile phase comprising acetonitrile. In some embodiments, the mobile phase gradient comprises an increase of said first mobile phase of from 22.0% to 44.1% over 38.5 minutes. [00251] In some embodiments, detecting fluorescence comprises generating a chromatogram, wherein the dependent variable is selected from mobile phase volume, eluent volume passed through the chromatographic column, or time, and the observable is fluorescence. In certain embodiments, determining an N-glycan profile of a population of anti-CD20 antibody proteins comprises quantifying the relative amounts of labeled G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans of said population. In certain embodiments, chromatography (i.e., HILIC-UPLC) is performed using a flow rate, column temperature, mobile phase gradient, and period of time parameters sufficient to separate the N-glycans of G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F for quantification. In certain embodiments, the amount of an N- glycan is quantified by calculating its area under a curve comprising labeled G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans in a chromatograph separating said N-glycans. In certain embodiments, the relative abundance of an N-glycan selected from G0-GN, G0F-GN, GO, Gl-GN, GOB, G0F, Man5, G0FB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, or G2F is quantified by calculating the percent peak area of said N- glycan relative to total peak area of GO-GN, GOF-GN, GO, Gl-GN, GOB, GOF, Man5, GOFB, Gl, Gl', GIB, GIF, GIF', G1FB, G2, and G2F N-glycans in a chromatograph separating said N- glycans. In certain embodiments, the peak area of an N-glycan in the chromatograph is greater than or equal to 0.25%. In certain embodiments, the peak area of an N-glycan in the chromatograph has a signal-to-noise ratio of greater than or equal to 3.0.
[00252] An N-glycan profile of a population of anti-CD20 antibody proteins may be determining the relative amounts of glycosylation of the population via a liquid chromatography- coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific glycosylation results of a digested population of anti-CD20 antibody proteins.
[00253] In some embodiments, the LC-MS peptide mapping comprises determining the molecular weight of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of glycosylated residues (i.e., GO, GOF, GOB, GOFB, Gl, or GIF). In some embodiments, the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion. In certain embodiments, the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest. In certain embodiments, the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
[00254] In some embodiments, the observed molecular weights are compared to theoretical masses for glycosylated peptides selected from Table 1, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 1, column 4. Table 1: Peptides for determining glycosylation following Lys-C digestion
Figure imgf000058_0001
[00255] In some embodiments, the population of anti-CD20 antibody proteins comprises glycosylation at residue Asn-298 of SEQ ID NO: 33 present in a heavy chain.
(c) Methods of Determining Deamidation
(i) Deamidation In General
[00256] Also provided herein is a method of determining the amount of asparagine deamidation in a population of anti-CD20 antibody proteins. In some embodiments, the amount of asparagine deamidation in a population of anti-CD20 antibody proteins is the amount of isoaspartate residues in the population. In some embodiments, the method comprises detecting the amount of isoaspartate residues in a digested population of anti-CD20 antibody proteins.
(ii) Deamidation By Isoquant
[00257] The amount of asparagine deamidation or isoaspartate in a population or digested population of anti-CD20 antibody proteins may determined via enzymatic method, for example, by the addition of a methyltransferase (i.e., protein isoaspartyl methyltransferase (PIMT)), which catalyzes the transfer of a methyl group from S-adenosyl methionine (SAM) to isoaspartic acid. This PIMT- catalyzed reaction produces S-adenosyl homocysteine (SAH) in an amount that is directly proportional (~1 : 1 stoichiometry) to the level of isoaspartate present in a sample. SAH can be directly measured by separating SAH (i.e., via ultra performance liquid chromatography (UPLC) or reversed-phase high performance liquid chromatography (RP-HPLC)), detecting at 260 nm, and quantifying the amount of SAH by comparing to a SAH standard curve (i.e., Trp- Ala-Gly-Gly-isoAsp-Ala-Ser-Gly-Glu peptide).
[00258] In some embodiments, the method comprises a step of derivatization comprising contacting the population of anti-CD20 antibody proteins with PIMT and SAM. In some embodiments, the population of anti-CD20 antibody proteins is contacted with PIMT and SAM at a temperature and for a period of time sufficient for derivatization (i.e., methylation) of substantially all of the aspartate residues present in the population. In some embodiments, the method optionally comprises a step of quenching the derivatization reaction. In certain embodiments, the method comprises a step of separating SAH by hydrophobicity for quantification of isoaspartate. In some embodiments, separating SAH comprises chromatography, which comprises a hydrophobic interaction stationary phase and a reversed- phase eluent. In some embodiments, separating SAH comprises chromatography using a Cl 8 stationary phase. In some embodiments, the Cl 8 stationary phase is a Hydro-RP column. In certain embodiments, the revers ed-phase eluent comprises one or more mobile phase (i.e., a weakly acidic phosphate buffer and/or methanol). In certain embodiments, chromatography is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate SAH from the derivatization reaction components. In some embodiments, chromatography is performed using a chromatography system that controls these parameters (i.e., an HPLC or UPLC system). In certain embodiments, the chromatography system is coupled to a detector, for detecting SAH after derivatization (i.e., absorbance at 260 nm). In some embodiments, separation and detection of SAM is performed using a hydrophilic interaction stationary phase, a reversed-phase eluent, and a RP-HPLC or UPLC system equipped with detector for absorbance of SAH at about 260 nm. In some embodiments, the hydrophobic interaction stationary phase comprises a Synergi Hydro-RP (4.6 mm X 150 mm). In some embodiments, the stationary phase (i.e., column) temperature is about 25 °C or room temperature. In specific embodiments, the flow rate is 1 mL/min. In some embodiments, the reversed-phase eluent comprises one or more mobile phases comprising a first mobile phase comprising about 50 mM potassium phosphate, at a pH of about 6.2, and a second mobile phase comprising methanol. In some embodiments, the mobile phase gradient comprises said second mobile phase at 10.0% at 0 minutes, 40% at 7.5 minutes, 80% at 10.5 minutes, 80% at 12.5 minutes, 10% at 13.5 minutes, 10% at 20 minutes, and 10% at 25 minutes. In some embodiments, the amount of SAH is quantified by comparing its integrated curve area to a SAH standard curve. In some embodiments, the SAH standard curve is prepared using Trp-Ala-Gly- Gly-isoAsp-Ala-Ser-Gly-Glu. In some embodiments, the amount of asparagine deamidation or isoaspartate in a population of anti-CD20 antibody proteins is determined using the Promega Isoquant kit along with Isoasp-DSIP standard, in accordance to manufacturer’s protocol.
(iii) Deamidation by LC-MS
[00259] The amount of asparagine deamidation or isoaspartate in a population or digested population of anti-CD20 antibody proteins may determined via a liquid chromatography-coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific deamidation results of a digested population of anti-CD20 antibody proteins. [00260] In some embodiments, the LC-MS peptide mapping comprises determining the molecular weight and/or relative abundance of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of deamidation or isoaspartate residues. In some embodiments, the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion. In certain embodiments, the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest. In certain embodiments, the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
[00261] In some embodiments, the observed molecular weights are compared to theoretical masses for deamidated peptides selected from Table 2, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 2, column 3. Table 2: Peptides for determining deamidation following Lys-C digestion
Figure imgf000061_0001
[00262] In some embodiments, the LC-MS peptide mapping comprises determining the molecular weight and/or relative abundance of peptides derived from an Asp-N digested population of anti-CD20 antibody proteins to determine the presence and amount of deamidation or isoaspartate residues. In some embodiments, the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion. In certain embodiments, the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Asp-N digest. In certain embodiments, the relative abundance of a peptide of an Asp-N digest is calculated by integrating its area under the curve compared to total area.
[00263] In certain embodiments, the observed molecular weights are compared to theoretical masses for deamidated peptides selected from Table 3, column 2. In certain embodiments, the observed molecular weights are compared to theoretical masses selected from Table 3, column 3.
Table 3: Peptides for determining deamidation following Asn-N digestion
Figure imgf000062_0001
(d) Method of Determining Oxidation
(i) Oxidation In General
[00264] Also provided here is a method of determining the level of methionine oxidation in a population of anti-CD20 antibody proteins as described herein. In some embodiments, the amount of methionine oxidation in a population of anti-CD20 antibody proteins is the amount of Met sulfoxide (MetO) residues in the population. In some embodiments, the method comprises detecting the amount of Met sulfoxide (MetO) residues in the population.
(ii) Oxidation by LC-MS
[00265] The amount of methionine oxidation or Met sulfoxide (MetO) residues in a population or digested population of anti-CD20 antibody proteins may be determined via a liquid chromatography-coupled mass spectrometer (LC-MS) peptide mapping method, for example, by summing site specific oxidation results of a digested population of anti-CD20 antibody proteins. [00266] In some embodiments, the LC-MS peptide mapping comprises determining the molecular weight of peptides derived from a Lys-C digested population of anti-CD20 antibody proteins to determine the presence and amount of oxidation or MetO residues. In some embodiments, the population of anti-CD20 antibody proteins is reduced and alkylated prior to digestion. In some embodiments, the LC is performed using parameters for flow rate, stationary phase (i.e., column) temperature, mobile phase gradient, and period of time, wherein the parameters in combination are sufficient to separate the peptides of the Lys-C digest. In some embodiments, the relative abundance of a peptide of a Lys-C digest is calculated by integrating its area under the curve compared to total area.
[00267] In some embodiments, the observed molecular weights are compared to theoretical masses for oxidized peptides selected from Table 4, column 2. In some embodiments, the observed molecular weights are compared to theoretical masses selected from Table 4, column 3.
Table 4: Peptides for determining oxidation following Lys-C digestion
Figure imgf000063_0001
[00268] In some embodiments, the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a heavy chain. In some embodiments, the one or more methionine residues present in the heavy chain are selected from Met-20, Met-34, Met-81, Met-253 or Met-428 as shown in SEQ ID NO: 17, 18, 21, 24, or 29, respectively. In certain embodiments, the population of anti-CD20 antibody proteins comprises methionine oxidation at one or more methionine residues present in a light chain. In some embodiments, the one or more methionine residues present in the light chain are selected from Met-21 or Met-32 as shown in SEQ ID NO:30.
(e) Assay for Determining Protein Conformation— Circular dichroism
[00269] In some embodiments, the secondary structures of anti-CD20 antibody proteins are analyzed using Circular Dichroism (CD) spectroscopy by measuring the difference in absorption between left and right circularly polarized light due to structural asymmetry. The CD spectroscopy using far-ultraviolet spectra with a wavelength between approximately 170 and 260 nm. At these wavelengths, the different secondary structures commonly found in protein can be analyzed, since a-helix, parallel and anti-parallel |3-sheet, |3-Turn, and random coil conformations each give rise to a characteristic spectra, the spectrum of a given protein can be used to estimate its percentage content on the secondary structures.
7.6 Single Batch Compositions
[00270] Provided herein are compositions or populations of anti-CD20 antibody proteins described in Section 7.4 at scaled up quantities. In certain embodiments, these scaled up quantities are present in a single batch, ie, a composition derived from a single run from a single bioreactor of a specified volume. For example, the anti-CD20 antibody proteins obtained from a single run of a 15,000L bioreactor can be referred to as a single batch. In certain embodiments, the bioreactor has a capacity of at least 100; 200; 300; 400; 500; 750; 1,000; 2,000; 3,000; 4,000; 5,000; 7,500; 10,000; 15,000; 20,000; or at least 25,000 L. In certain embodiments, anti-CD20 antibody proteins are present in such a single batch at a concentration of at least 10 mg/ml;
15mg/ml; 20mg/ml; 25mg/ml; or at least 30mg/ml as determined using an assay described in Section 7.6(a). In certain embodiments, anti-CD20 antibody proteins are present in such a single batch at a concentration of at between 10 to 35 mg/ml; 10 to 30 mg/ml; 10 to 25 mg/ml; 10 to 20 mg/ml; 10 to 15 mg/ml; 15 to 35 mg/ml; 15 to 30 mg/ml; 15 to 25 mg/ml; 15 to 20 mg/ml; 20 to 35 mg/ml; 20 to 30 mg/ml; 20 to 25 mg/ml; 25 to 35 mg/ml; or 25 to 30 mg/ml as determined using an assay described in Section 7.6(a). In certain embodiments, anti-CD20 antibody proteins are present in such a single batch at a concentration of about 15mg/ml; about 20mg/ml; about about 25mg/ml; about 30mg/ml; or about 35mg/ml as determined using an assay described in Section 7.6(a) (wherein “about” means +/- 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%).
[00271] In some aspects, the anti-CD20 antibody proteins disclosed herein are produced at a commercial scale. In certain embodiments, the commercial scale is 10,000 to 25,000 L.
(a) Total protein
[00272] In some embodiments, the amount of total protein is measured using spectrophotometry. In certain embodiments, the amount of total protein is measured by absorbance at 280 nm.
[00273] In some embodiments, the amount of total protein can be measured using the following assay procedures. This method determines the concentration of the anti-CD20 antibody proteins using an ultraviolet absorbance measurement at 280 nm with a light scattering correction at 320 nm. The calculation of concentration is based on the Beer-Lambert law. The extinction coefficient of the anti-CD20 antibody protein at 280 nm is 1.61 mL/mg/cm as determined by the amino acid composition. The extinction coefficient can be used. The method is suitable for use for post Protein A purification in-process sample testing, release testing, and stability testing. Test samples are diluted gravimetrically in triplicate with 0.9% sodium chloride to a target protein concentration of 0.4 mg/mL. Saline is used as a blank for the instrument. Bovine serum albumin is used as a system suitability control and is tested before and after sample measurements. Test samples are loaded into a quartz cuvette with a 1 cm pathlength and the absorbances at 280 and 320 nm are measured. The test samples are tested in triplicate. In certain embodiments, the amount of total protein can be measured using the critical material and equipment listed in Table 5. In certain embodiments, the amount of total protein can be measured using the protein concentration system suitability criteria listed in Table 6.
Table 5: Critical Material and Equipment Used in the Protein Concentration Assay
Figure imgf000065_0001
UV = ultraviolet; Vis = visible light
Table 6: Protein Concentration System Suitability Criteria
Figure imgf000065_0002
AU = absorbance unit; CV = coefficient of variation
[00274] In some embodiments, the population of anti-CD20 antibody proteins provided herein has an amount of total protein of 25.5-25.8 mg/mL. In certain embodiments, the population has an amount of total protein of about 25.6 mg/mL.
7.7 Biological Properties
[00275] In some embodiments, the populations of anti-CD20 antibody proteins provided herein possess biological properties detailed in this section. In certain embodiments, the biological properties can be measured using the assays described in Section 7.7(a). The biological properties of the anti-CD20 antibody protein compositions provided herein are described in Section 7.7(b).
(a) Assays
(i) Cell-based antibody-dependent cellular cytotoxicity (ADCC)
[00276] In some embodiments, the cell-based ADCC assay uses Raji cells as target cells. In certain embodiments, the Raji cells express CD20. In certain embodiments, the cell-based ADCC assay uses effector cells selected from CD 16 effector cells and primary NK cells. In certain embodiments, the cell-based ADCC assay uses Raji cells as target cells and KILR CD16a effector cells effector cells. In certain embodiments, the cell-based ADCC assay uses Raji cells as target cells and primary NK cells as effector cells. In certain embodiments, the KILR CD 16a effector cells are single donor-derived human CD 8+ T-lymphocytes engineered to express CD16/FcyRIII on their plasma membrane surface.
[00277] In aome embodiments, the cell-based ADCC assay uses target cell lysis as the read out. In some embodiments, the target cell lysis is quantified using CytoTox Gio™ (Promega). In some embodiments, the cell-based ADCC assay shows relative potency of the population against a commercial reference standard. In some embodiments, the cell-based ADCC assay produces a dose-response curve and an EC50 value. In some embodiments, a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells at more than 100% of that of commercial reference standard RS-117808.
[00278] In some embodiments, the ADCC activity can be measured using a CD 16 activity assay. In some embodiments, the CD 16 activity assay assesses ADCC activity using a surrogate read-out. In some embodiments, the CD 16 activity assay uses WIL2-S as target cells. In certain embodiments, the effector cells used are a stably transformed Jurkat cell line expressing a chimeric molecule comprising of the extracellular domain of FcyRIIIa joined to the transmembrane and intracellular domains of the gamma chain of the mast cell/basophil Fc receptor for IgE. In certain embodiments, cells are combined and treated with a serial dilution of the anti-CD20 antibody in the presence of PMA (Phorbol 12- Myristate 13-Acetate). In certain embodiments, the activation of the effector cells induces the release of IL-2, which is measured by a commercial ELISA kit. In certain embodiments, the potency is reported as a percentage relative to a reference standard. [00279] In some embodiments, the population has a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard. In some embodiments, the population has a relative potency of about 115% in a CD 16 activity assay compared to a commercial reference standard. In some embodiments, the commercial reference standard is RS- 117808.
[00280] In some embodiments, the cell-based ADCC assay uses the following assay procedures. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are used in the concentration range of 250.00 pg/mL to 0.04 pg/mL (250, 50, 16.7, 5.6, 1.9, 0.6, 0.2, 0.04 pg/mL). Two independent preparations of the materials are prepared and assayed across duplicate plates. Assay controls are prepared in triplicates and include: Target Cells Alone Control, Target Cells Death Control, Effector Cells Alone Control, and Effector & Target Cells Control. KILR cells are obtained from Eurofins. They come from a master cell bank made from a unique human donor. A Master and Working cell bank system and qualification protocol allows the vendor to create many thousands of vials in a reproducible manner and TG maintains a secure supply of this critical reagent. Procedures, such as incoming material qualification process, provide assurance that highly reproducible cells are used in every assay. KILR cells are thawed and cultured at 1x10A6 cells/mL in media supplemented with IL-2, and are rested for a minimum of 6 days before use and are not be used beyond 14 days post thaw. To perform the KILR ADCC assay, Raji cells, also managed through a master and working cell banking system, are seeded at 1x105 cells/mL, and reference standard, internal assay control, and testing samples dilutions are added to the Raji cells. KILR Effector cells are seeded at 5x105 cells/mL to a final effector: target (E:T) ratio of 5: 1. Cells are cultured at 36±1°C, 5±1%CO2 for 18 to 22 hours. At the end of the incubation, a CytoTox GLo™ preparation is added, and plates are incubated for 30±10 minutes. The plates are read using the SpectraMax plate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data is evaluated using the SoftMax Pro software for potency against the reference standard. Representative reference standard and sample dose response curves are shown. Results are reported as percentage potency relative to the primary reference standard or its derivative.
[00281] In some embodiments, the cell-based ADCC assay is used as a potency assay for batch release. In some embodiments, the cell-based ADCC assay is used as a potency assay for stability testing. In some embodiments, the cell-based ADCC assay is used as a potency assay in the manufacturing quality controls and processes. In some embodiments, the cell-based ADCC assay is used in comparison studies (for example, in research and development or clinical studies).
[00282] In some embodiments, the higher potency of the population in a cell-based ADCC assay is related to the lower level of fucose content in the N-glycan profile of the population. In some embodiments, the higher potency of the population in a cell-based ADCC assay is due to the lower level of fucose content in the N-glycan profile of the population.
(ii) Antibody-dependent cellular phagocytosis (ADCP)
[00283] In some embodiments, the antibody-dependent cellular phagocytosis (ADCP) activity can be measured using CD20 expressing Daudi cells as target cells (labeled by PKH26). In some embodiments, human monocytes are isolated from PBMC and differentiated in vitro using GM- CSF to yield macrophages. In some embodiments, macrophages are co-cultured with PKH26- labeled target cells previously incubated with serial diluted anti-CD20 antibody samples. In some embodiments, target cell phagocytosis was assessed by flow cytometry. In some embodiments, the dose response curves, as well as calculated IC50s of the samples tested can be shown.
(iii) Complement dependent cytotoxicity (CDC)
[00284] In some embodiments, the cell-based CDC assay uses Jeko-1 cells as target cells and rabbit compliment system. In certain embodiments, the cell-based CDC assay uses Raji cells as target cells and human complement system. In some embodiments, the cell-based CDC assay uses target cell lysis as the read out.
[00285] In some embodiments, the cell-based CDC assay uses the following assay procedures. Nine-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are used in the concentration range of 10,000 ng/mL to 10.42 ng/mL (10,000.00, 3333.33, 1666.67, 833.33, 416.67, 208.33, 104.17, 52.08, 10.42 ng/mL). Two independent preparations of the materials are prepared and assayed across duplicate plates. Assay negative controls are prepared in triplicates and include target cells and complement control and target cells alone control. In this assay, Jeko-1 cells, obtained from the ATCC and maintained through a master banking system, are seeded at 3x105 cells/mL and incubated for 60-90 minutes. Reference standard, internal assay control, and test samples diluted in duplicate in cell culture medium are added. Complement is then added to the wells and the plates are incubated for approximately 2 hours at 36°C±1 °C followed by 25±5 minutes at room temperature. Controls include a target cells with complement only control and a target cell only control to provide a basal level of target cell viability over the course of the assay. The Cell Titer-Gio reagent is then added and incubated an additional 30±10 minutes at room temperature. At the end of the assay, the plates are read using a SpectraMax M5 plate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data are evaluated using the Softmax Pro software for potency against the reference standard. Results are reported as % potency relative to the reference standard. Representative reference standard and sample dose response curves are shown.
(iv) CD20 binding activity
[00286] In some embodiments, the cell-based CD20 binding assay uses Jeko-1 cells and an MSD assays. In some embodiments, the cell-based CD20 binding assay produces a dose dependent binding curve and a EC50 value.
[00287] In some embodiments, the cell-based CD20 binding assay uses the following assay procedures. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are prepared in the concentration range of 40,000.00 ug/mL to 0.23 ng/mL (40,000.00, 4,000.00, 1,000.00, 333.30, 111.10, 37.00, 4.60, 0.23 ng/mL). Two independent preparations of the Test Material are prepared for each 2-plate Test Material assessment. Assay controls include No cell control (Reference Standard/Test Material dilution + detection reagent, omitting cells) and Cell only control (Cells + detection reagent, omitting Reference Standard/Test Material). Jeko-1 cells are seeded onto MSD high bind plate in PBS at 3 x 105 cells per mL, in a final volume of 100 pL per well, and incubated at 35 to 37°C for 2 hours ±10 minutes. Unbound cells are removed by a PBS wash, the plates are blocked with 45% FBS. Fifty pL of Reference Standard, QC Reference Control, or Test Materials dilutions are added, and the plate is incubated at room temperature for 1 hour ±10 min while shaking. Following incubation and three PBS washes, 50 pL anti-human Fc detection antibody conjugated with STREP- SULFOTAG is added and incubated for 1 hour ± 5 minutes at room temperature while shaking. The plates are washed again with PBS and 150 pL of the MesoScale read buffer, containing tripropylamine (TP A), is added as a co-reactant for light generation for an electrochemiluminescence read out. Plates are read immediately on a MSD Reader using Workbench 4.0. The resulting data are evaluated using the PLA software and analyzed using a constrained 4 parameter logistic model to generate a relative binding, 95% confidence intervals and results relative to the reference standard. Binding activity results are reported as percentage potency relative to the reference standard. Representative reference standard and testing sample dose response curves, from which relative potency test result is calculated.
(v) FcyRIIIa 158V and FcyRIIIa 158F binding activity
[00288] In some embodiments, the FcyRIIIa binding assay uses Surface Plasmon Resonance (SPR). In certain embodiments, the FcyRIIIa binding assay produces sensorgrams showing a dose dependent binding, saturation, and dissociation. In certain embodiments, the FcyRIIIa binding assay calculates dissociation constant by the on and off rate and steady state kinetics. In certain embodiments, the binding affinity to FcyRIIIa 158V of the population is approximately one order of magnitude higher than binding affinity to FcyRIIIa 158F of the population. In certain embodiments, the binding affinity to FcyRIIIa 158V of the population is significantly higher than Rituxan. In certain embodiments, the binding affinity to FcyRIIIa 158F of the population is significantly higher than Rituxan. In certain embodiments, the binding affinities of the population to both FcyRIIIa 158V and FcyRIIIa 158F are significantly higher than Rituxan. [00289] In some embodiments, the FcyRIIIa binding assay uses the following assay procedure. FcyRIIIa 158V receptor (1.2 pg/ml) is immobilized on the chip surface using covalent amine coupling chemistry. Eight-point dilution series of the Reference Standard, QC Reference Control, and Test Materials are prepared in the concentration range of 2000 nM to 15.6 nM with a dilution factor of 2. Independent duplicates of sample dilutions are injected over the chip, followed by surface regeneration between each cycle. The binding is measured in response units (RU). The kinetics of the binding reaction is determined by measuring changes in SPR due to the increase in mass in the close proximity to the biosensor chip surface. Change in the mass of the complex as a function of time is visualized as a sensorgram. The equilibrium dissociation constants (KD) and the relative affinities of each sample relative to the Reference Standard are determined for each receptor. The rates of change of the SPR signal are analyzed using a 1 : 1 Langmuir model for FcyRIIIa 158V variant to yield apparent rate constants for the association and dissociation phases of the reaction, and equilibrium dissociation constants. KD is determined using steady state affinity for the FcyRIIIa 158F variant. The binding signals are exported into PLA to determine the relative binding response. Results are reported as % potency relative to the reference standard for each FcyRIIIa variant, 158V and 158F.
(vi) Cl q binding activity
[00290] In some embodiments, the Clq binding activity can be as measured using ELISA. In certain embodiments, the Clq binding activity can be as measured using the following assay procedure. A 7-point dilution series of the reference standard, QC reference control, and test materials are prepared in the concentration range of 15.00 pg/mL to 0.12 pg/mL. Reference standard, QC reference control, and test materials dilutions are coated onto an ELISA plates, and the plates incubated for 1 hour ± 30 minutes at room temperature (shaking 150 to 200 rpm). After coating, the plates are washed (3x with PBS/0.05% Tween), blocked (with 1% BSA and incubated for 1 hour ± 10 minutes at room temperature and shaking 150 to 200 rpm), and washed (3x with PBS/0.05% Tween). Clq that has been conjugated with peroxidase is then added, and the plates incubated for 1.5 hours ± 30 minutes at room temperature (shaking 150 to 200 rpm). After incubation and washing, a tetramethylbenzidine (TMB) substrate solution is added and plates incubated at room temperature for 7 minutes (-1 minute/±30 seconds). This produces a colorimetric reaction which is proportional to the level of Clq bound. The reaction is stopped with the addition of IM sulfuric acid and the color is measured at 450 nm using a Molecular Devices SpectraMax microplate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data are evaluated using the SoftMax Pro software for potency against the reference standard. Clq binding activity results are reported as percentage potency relative to the reference standard. Representative dose response curves from one assay can be shown.
(vii) B-cell depletion activity
[00291] In some embodiments, the B-cell depletion activity can be measured in a human whole blood B-cell depletion assay. In certain embodiments, the B-cell depletion activity can be measured in an autologous normal human whole blood B-cell depletion assay. In certain embodiments, the B-cell depletion can be measured by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3 -positive T cells, CD 19-positive B-cells, and CD20- positive B-cells. In certain embodiments, percent of B-cell depletion (100-([100/B-/T-cell ratio in sample without antibody] x [B-/T-cell ratio in sample containing antibody])) can be calculated and plotted against sample concentration. [00292] In some embodiments, the B-cell depletion activity can be measured using blood from three healthy donor(s). In certain embodiments, the B-cell depletion can be measured by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3-positive T cells, CD 19-positive B-cells, and CD20-positive B-cells.
(b) Biological Properties
[00293] In some embodiments, the biological properties of a composition or population of anti-CD20 antibody proteins provided herein can be measured and described in an assay described in Section 7.7(a) and with the use of a comparison with a reference standard. In certain embodiments, the reference standard is a commercial reference standard.
[00294] In some embodiments, the reference standard is an anti-CD20 antibody. In certain embodiments, the reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
[00295] In some embodiments, the commercial reference standard is RS-117808. In certain embodiments, the populations of anti-CD20 antibody proteins provided herein possess the biological properties as shown in Table 7. RS-117808 (“antibody Ublituximab (TG-1101)”) was deposited according to the terms of the Budapest Treaty at the American Type Culture Collection (ATCC), located at 10801 University Boulevard, Manassas, VA 20110, received by the ATCC on April 15, 2022, and assigned unofficial patent deposit number PTA-127294.
Table 7: Biological Properties in Comparison to Reference Standard
Figure imgf000072_0001
(i) Cell-based antibody-dependent cellular cytotoxicity (ADCC)
[00296] In some embodiments, a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells assay (see Section 7.7(a)(i)) with are relevant potency that is at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, or at at least 200% of that of the commercial reference standard RS-117808. In certain embodiments, a composition or population of anti-CD20 antibody proteins provided herein performs in a cell-based ADCC using CD 16 effector cells assay (see Section 7.7(a)(i)) with are relevant potency that is at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150- fold, or at least 200-fold of that of the commercially available Rituxan® (Genentech/Biogen). [00297] In some embodiments, the population induces cytotoxicity in a cell-based antibodydependent cellular cytotoxicity (ADCC) assay. In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to a commercial reference standard.
[00298] In some embodiments, the population induces greater cytotoxicity in an ADCC assay compared to an anti-CD20 antibody. In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces greater cytotoxicity in an ADCC assay compared to OCREVUS (ocrelizumab).
[00299] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a cell-based ADCC assay compared to a commercial reference standard.
[00300] In some embodiments, the population has a relative potency of between at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50- fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300- fold in a cell-based ADCC assay compared to a commercial reference standard.
[00301] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative potency of about 38% to about 3 -fold in a cell-based ADCC assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population has a relative potency of about 10-fold to about 31 -fold in a cell-based ADCC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 28-fold to about 2250-fold in a cell-based ADCC assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 25-fold in a cell-based ADCC assay compared to OCREVUS (ocrelizumab).
[00302] In some embodiments, the population has a relative potency of 60 to 200%, 70 to 190%, 80% to 180%, 85 to 170%, or 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of about 117% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
[00303] In some embodiments, the EC50 value for the ADCC potency as measured in a cellbased ADCC assay of the population is between 2 and 6 pg/mL. In certain embodiments, the EC50 value for the ADCC potency as measured in a cell-based ADCC assay of the population is between 0.2 and 20 pg/mL, 0.3 and 18 pg/mL, 0.4 and 15 pg/mL, 0.5 and 12 pg/mL, 0.6 and 10 pg/mL, 0.7 and 9 pg/mL, 0.8 and 8 pg/mL, 0.9 and 7 pg/mL, or 1 and 6 pg/mL. In certain embodiments, the EC50 value for the ADCC potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty or more repeated cell-based ADCC assays. In certain embodiments, the EC50 value for the ADCC potency as measured in a cell-based ADCC assay (e.g., using the Eurofins/DiscoverX KILR CD16a effector cells) of the population is about 5.45 pg/mL. In certain embodiments, the EC50 value for the ADCC potency as measured in a cell-based ADCC assay (e.g., using the Eurofins/DiscoverX KILR CD16a effector cells) of the population is about 2.42 pg/mL.
(ii) Antibody-dependent cellular phagocytosis (ADCP)
[00304] In some embodiments, the population induces antibody-dependent cellular phagocytosis (ADCP). In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to an anti-CD20 antibody. [00305] In some embodiments, the population induces greater phagocytosis in an ADCP assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population induces phagocytosis in an ADCP assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater phagocytosis in an ADCP assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the
1 population induces greater phagocytosis in an ADCP assay compared to OCREVUS (ocrelizumab).
[00306] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in an ADCP assay compared to a commercial reference standard.
[00307] In some embodiments, the population has a relative potency of between at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150- fold, 200-fold or at most 300-fold in an ADCP assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100- fold, 100-fold and 150-fold, or between 150-fold and 300-fold in an ADCP assay compared to a commercial reference standard.
[00308] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative potency of about 7-fold in an ADCP assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 15-fold in an ADCP assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 23 -fold in an ADCP assay compared to GAZYVA (obinutuzumab).
[00309] In some embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 0.1 and 1 ng/mL. In certain embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 1 and 10, 2 and 9, 3 and 8, 4 and 7, or 5 and 6 ng/mL. In certain embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is between 0.05 and 20 ng/mL, 0.1 and 19 ng/mL, 0.15 and 18 ng/mL, 0.2 and 18 ng/mL, 0.25 and 15 ng/mL, 0.3 and 12 ng/mL, 0.3 and 10 ng/mL, 0.3 and 9 ng/mL, 0.3 and 8 ng/mL, 0.3 and 7 ng/mL, or 0.3 and 6 ng/mL. In certain embodiments, the EC50 value for the ADCP potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated ADCP assays. In certain embodiments, the EC50 value for the ADCP potency as measured in an ADCP assay of the population is about 5.50 ng/mL.
(iii) Complement dependent cytotoxicity (CDC)
[00310] In some embodiments, the population induces complement dependent cytotoxicity (CDC). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to a commercial reference standard. In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to an anti-CD20 antibody.
[00311] In some embodiments, the population induces greater cytotoxicity in a CDC assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to RITUXAN (rituximab). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to veltuzumab (IMMU-106). In certain embodiments, the population induces greater cytotoxicity in a CDC assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces greater cytotoxicity in a CDC assay assay compared to OCREVUS (ocrelizumab).
[00312] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a cell-based CDC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200- fold in a cell-based CDC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in a cell-based CDC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a cell-based CDC assay compared to a commercial reference standard.
[00313] In some embodiments, the population has a relative potency of between at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a cell-based CDC assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a cell-based CDC assay compared to a commercial reference standard. [00314] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative potency of about 50% in a cell-based CDC assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 37% in a cell-based CDC assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 1.8-fold in a cell-based CDC assay compared to OCREVUS (ocrelizumab).
[00315] In some embodiments, the population has a relative potency of 50 to 150%, 60 to 140%, 70 to 130%, 75 to 120%, or 78 to 116% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 78 to 116% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 73 to 128% or 74 to 127% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of about 91% in a cell-based CDC assay compared to that of a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808. [00316] In some embodiments, the EC50 value for the CDC potency as measured in a cellbased CDC assay of the population is between 0.4 and 0.7 pg/mL or between 0.4 and 0.6 pg/mL. In certain embodiments, the EC50 value for the CDC potency as measured in a cell-based CDC assay of the population is between 0.05 and 5 pg/mL, 0.1 and 4 pg/mL, 0.15 and 3 pg/mL, 0.2 and 2 pg/mL, 0.25 and 1 pg/mL, 0.3 and 0.9 pg/mL, 0.3 and 0.8 pg/mL, or 0.3 and 0.7 pg/mL. In certain embodiments, the EC50 value for the CDC potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated cell-based CDC assays. In certain embodiments, the EC50 value for the CDC potency as measured in a cell-based CDC assay of the population is about 0.5 pg/mL.
(iv) CD20 binding activity
[00317] In some embodiments, the population possesses CD20 binding activity in a cell-based CD20 binding assay. In certain embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to a commercial reference standard. [00318] In some embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population possesses greater CD20 binding activity compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater CD20 binding activity compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater CD20 binding activity compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater CD20 binding activity compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater CD20 binding activity compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces possesses greater CD20 binding activity compared to OCREVUS (ocrelizumab).
[00319] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a CD20 binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a CD20 binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in a CD20 binding compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a CD20 binding compared to a commercial reference standard.
[00320] In some embodiments, the population has a relative potency of between at least 5%,
10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a CD20 binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a CD20 binding assay compared to a commercial reference standard. [00321] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative potency of about 1.6-fold to about 5.8-fold in a CD20 binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population has a relative potency of about 81% to about 4.3 -fold in a CD20 binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 1.6-fold to about 4.1 -fold in a CD20 binding assay compared to RITUXAN (rituximab).
[00322] In some embodiments, the population has a relative potency of 50 to 150%, 60 to 140%, 70 to 130%, 80 to 120%, 90 to 120%, or 92 to 118% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 92 to 118% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of 82 to 138% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the population has a relative potency of about 109% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
[00323] In some embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is between 0.05 and 0.1 pg/mL. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is between 0.01 and 0.5 pg/mL, 0.02 and 0.4 pg/mL, 0.03 and 0.3 pg/mL, 0.04 and 0.2 pg/mL, or 0.05 and 0.1 pg/mL. In certain embodiments, the EC50 value for the CD20 binding potency is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated cell-based CD20 binding activity bioassays. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is about 0.093 pg/mL. In certain embodiments, the EC50 value for the CD20 binding potency as measured in a cell-based CD20 binding activity bioassay of the population is about 0.063 pg/mL.
(v) FcyRIIIa 158V and FcyRIIIa 158F binding activity
[00324] In some embodiments, the population possesses FcyRIIIa 158V binding activity in an FcyRIIIa binding assay. In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to a commercial reference standard.
[00325] In some embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to ZE VALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater FcyRIIIa 158V binding activity in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab).
[00326] In some embodiments, the population has a relative FcyRIIIa 158V binding activity of at least 1000%, 750%, 500%, 250%, 100%, 75%, or at least 50% in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of at least more than 100% in a cell-based ADCC assay compared to a commercial reference standard. In certain embodiments, the population has a relative relative FcyRIIIa 158V binding activity of more than 100% in an FcyRIIIa binding assay compared to a commercial reference standard.
[00327] In some embodiments, the population has a relative FcyRIIIa 158V binding activity of at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100- fold, 125-fold, 150-fold, 200-fold or at most 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of at least between 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300- fold in an FcyRIIIa binding assay compared to a commercial reference standard.
[00328] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 3.8-fold in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 25.6-fold in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 18.7-fold in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative FcyRIIIa 158V binding activity of about 16-fold in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab). [00329] In some embodiments, the population possesses FcyRIIIa 158F binding activity in a FcyRIIIa binding assay. In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater CD20 binding activity in a cell-based CD20 binding assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater FcyRIIIa 158F binding activity in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab).
[00330] In some embodiments, the population has a relative FcyRIIIa 158F binding activity of at least 1000%, 750%, 500%, 250%, 100%, 75%, or at least 50% in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of at least more than 100% in a cell-FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative relative FcyRIIIa 158F binding activity of more than 100% in an FcyRIIIa binding assay compared to a commercial reference standard.
[00331] In some embodiments, the population has a relative FcyRIIIa 158F binding activity of at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30- fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100- fold, 125-fold, 150-fold, 200-fold or at most 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of at least between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150- fold and 300-fold in an FcyRIIIa binding assay compared to a commercial reference standard. [00332] In certain embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 2.6-fold in an FcyRIIIa binding assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 21.8-fold in an FcyRIIIa binding assay compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 10.2-fold in an FcyRIIIa binding assay compared to RITUXAN (rituximab). In certain embodiments, the population has a relative FcyRIIIa 158F binding activity of about 9.9-fold in an FcyRIIIa binding assay compared to OCREVUS (ocrelizumab). In certain embodiments, the population has significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab.
[00333] In some embodiments, the population has a relative potency of 20 to 300%, 30 to 250%, 40% to 220%, or 50 to 200% in a cell-based FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 50 to 200% in a cell-based FcyRIIIa binding assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 76 to 130% or 82 to 130% in a cell-based FcyRIIIa 158V binding assay compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
[00334] In some embodiments, the population has a KD value 55 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 10 to 100 nM, 15 to 90 nM, 10 to 80 nM, or 30 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 30 to 70 nM in an FcyRIIIa-158V binding assay as measured by surface plasmon resonance. In certain embodiments, the KD value is the average KD value calculated from the KD values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated FcyRIIIa- 158V binding assays. In certain embodiments, the population has a KD value about 59 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value about 64.1 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance.
[00335] In some embodiments, the population has a KD value 600 to 800 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 100 to 2000 nM, 200 to 1800 nM, 300 to 1700 nM, 400 to 1600 nM, 500 to 1500 nM, 500 to 1200 nM, 600 to 1000 nM, or 600 to 800 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In certain embodiments, the KD value is the average KD value calculated from the KD values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated FcyRIIIa- 158V binding assays. In certain embodiments, the population has a KD value 760 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance. In certain embodiments, the population has a KD value 680.3 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance.
(vi) Cl q binding activity
[00336] In some embodiments, the population possesses Clq binding activity as measured by ELISA. In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to a commercial reference standard.
[00337] In some embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, possesses greater Clq binding activity as measured by ELISA compared to GAZYVA (obinutuzumab). In certain embodiments, possesses greater Clq binding activity as measured by ELISA compared to ARZERRA (ofatumumab). In certain embodiments, possesses greater Clq binding activity as measured by ELISA compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater Clq binding activity as measured by ELISA compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population induces possesses possesses greater Clq binding activity as measured by ELISA compared to compared to OCREVUS (ocrelizumab).
[00338] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, or at least 25% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
[00339] In some embodiments, the population has a relative potency of between at least 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75- fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150- fold, 200-fold or at most 300-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a Clq binding assay as measured by ELISA compared to a commercial reference standard.
[00340] In some embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab). In certain embodiments, the population has a relative potency of about 142% in a Clq binding assay as measured by ELISA compared to ARZERRA (ofatumumab). In certain embodiments, the population has a relative potency of about 123% in a Clq binding assay as measured by ELISA compared to RITUXAN (rituximab). In certain embodiments, the population has a relative potency of about 112% in a Clq binding assay as measured by ELISA compared to OCREVUS (ocrelizumab).
[00341] In some embodiments, the population has a relative potency of 30 to 180%, 40 to 170%, 50 to 160%, 60 to 150%, 70 to 140%, 80 to 130%, 85% to 120%, or 88 to 113% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 88 to 113% or 86 to 117% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of 86 to 116% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the population has a relative potency of about 99% in a Clq binding assay as measured by ELISA compared to a commercial reference standard. In certain embodiments, the commercial reference standard is RS-117808.
[00342] In some embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is between 1.5 and 3 pg/mL. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is between 0.2 and 9 pg/mL, 0.3 and 8 pg/mL, 0.4 and 7 pg/mL, 0.5 and 6 pg/mL, 0.6 and 5 pg/mL, 0.7 and 4 pg/mL, 0.8 and 3 pg/mL, 0.9 and 2.9 pg/mL, or 1 and 2.8 pg/mL. In certain embodiments, the EC50 value for the Clq binding activity is the average EC50 value calculated from the EC50 values obtained in two, three, four, five, six, seven, eight, nine, ten, or more repeated ELISA experiments. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is about 1.92 pg/mL. In certain embodiments, the EC50 value for the Clq binding activity as measured by ELISA of the population is about 2.6 pg/mL. (vii) B cell depletion activity
[00343] In some embodiments, the population possesses B cell depletion activity as measured in a human whole blood B cell depletion assay. In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to a commercial reference standard.
[00344] In some embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to an anti-CD20 antibody. In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) and/or OCREVUS (ocrelizumab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to GAZYVA (obinutuzumab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to ARZERRA (ofatumumab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to RITUXAN (rituximab). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to veltuzumab (IMMU-106). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to ZEVALIN (ibritumomab tiuxetan). In certain embodiments, the population possesses greater B cell depletion activity as measured in a human whole blood B cell depletion assay compared to OCREVUS (ocrelizumab).
[00345] In some embodiments, the population has a relative potency of at least 1000%, 750%, 500%, 250%, 100%, 75%, 50%, 25%, 12%, or at least 5% in a human whole blood B cell depletion assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 5-fold, 10-fold, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125- fold, 150-fold, or at least 200-fold in a human whole blood B cell depletion assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of at least more than 100% in a human whole blood B cell depletion assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of more than 100% in a human whole blood B cell depletion assay compared to a commercial reference standard.
[00346] In some embodiments, the population has a relative potency of between at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750% 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or at least 200-fold and at most 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 500%, 750%, 1000%, 15-fold, 20-fold, 30-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, 200-fold or at most 300-fold in a human whole blood B cell depletion assay compared to a commercial reference standard. In certain embodiments, the population has a relative potency of between 5% and 50%, 50% and 100%, 100% and 500%, 500% and 1000%, 10-fold and 50-fold, 50-fold and 100-fold, 100-fold and 150-fold, or between 150-fold and 300-fold in a human whole blood B cell depletion assay compared to a commercial reference standard.
[00347] In certain embodiments, the commercial reference standard is an anti-CD20 antibody. In certain embodiments, the commercial reference standard is GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab).
7.8 Methods of Treating and Preventing Medical Conditions
Methods of Treating Multiple Sclerosis
[00348] Provided herein are methods of treating a relapsing form of multiple sclerosis (RMS) in a subject in need thereof, by administering to the subject an effective amount of an anti-CD20 antibody composition as described herein. In some embodiments, the relapsing form of multiple sclerosis (RMS) is selected from a clinically isolated syndrome (“CIS”), relapsing-remitting (“RRMS”), or active secondary progressive disease (“SPMS”). In some embodiments, the RMS is a CIS. In some embodiments, the RMS is RRMS. In some embodiments, the RMS is SPMS. In some embodiments, the subject is diagnosed with RMS in accordance to McDonald Criteria (2010), or by another suitable method known by one skilled in the art.
[00349] In some embodiments, the method comprises administering to the subject the anti- CD20 antibody composition as described herein in a multi-dosage regimen. In certain embodiments, the method comprises administering to the subject the anti-CD20 antibody composition as described herein by intravenous infusion. For example, anti-CD20 antibody composition described herein can be administered to the subject in a multi-infusion dosage regimen by intravenous infusion. Anti-CD20 antibody compositions described herein can be administered to the subject by intravenous infusion in a multi-infusion dosage regimen for 48 weeks. Additionally, or in the alternative, the anti-CD20 antibody composition described herein can be administered to the subject by intravenous infusion in a multi-infusion dosage regimen for 96 weeks. In some embodiments, the multi-infusion dosage regimen comprises first, second, and subsequent intravenous infusions of ublituximab. In some embodiments, “subsequent infusions” of ublituximab is any number of infusions after the second infusion.
[00350] In some embodiments, the intravenous infusion comprises a multi-dosage regimen (e.g., a multi-infusion dosage regimen), comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 2 weeks after the first infusion; c) a first subsequent infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition described herein (i.e., the anti- CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the first infusion; and d) one or more subsequent infusions comprising about 400 to about 500 mg of the anti-CD20 antibody composition described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the prior infusion.
[00351] In some embodiments, the intravenous infusion comprises a multi-dosage regimen (e.g., a multi-infusion dosage regimen), comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody composition at described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody composition described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody composition described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the prior infusion. In some embodiments, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; and subsequent infusions (e.g., the first subsequent infusion and/or the one or more subsequent infusions) are administered over 1 hour.
[00352] In some embodiments, the method comprises a first administration of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) by intravenous infusion to the subject and then a second administration of the anti-CD20 antibody as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) to the subject. In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising a prescribed dosage of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) herein at day 1; b) a second infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 2 weeks after the first infusion; c) a first subsequent infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti- CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the first infusion; and d) one or more subsequent infusions comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the prior infusion.
[00353] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising a prescribed dosage of an anti-CD20 antibody composition (e.g., GAZYVA (obinutuzumab), ARZERRA (ofatumumab), RITUXAN (rituximab), veltuzumab (IMMU-106), ZEVALIN (ibritumomab tiuxetan) or OCREVUS (ocrelizumab)) herein at day 1; b) a second infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti- CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti- CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at about 24 weeks or about six months after the prior infusion. In some embodiments, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; and subsequent infusions (e.g., the first subsequent infusion and/or the one or more subsequent infusions) are administered over 1 hour. [00354] In some embodiments, the method comprises a treatment period of at least 48 weeks. In some embodiments, the method comprises a treatment period of at least 96 weeks.
[00355] In some embodiments, the subject is a human. In some embodiments, the human subject is an adult. In some embodiments, the subject has experienced at least one relapse in the previous year prior to treatment or two relapses in the previous two years prior to treatment. In some embodiments, the subject has or had the presence of a T1 gadolinium (Gd)-enhancing lesion in the previous year prior to treatment with the anti-CD20 antibody composition as described herein. In some embodiments, the subject has an Expanded Disability Status Scale (EDSS) score of from 0 to 5.5 prior to treatment with the anti-CD20 antibody composition as described herein.
[00356] In some embodiments of the method, the subject has not been treated with a nonsteroid therapy for MS in the previous five years prior to treatment with the anti-CD20 antibody composition as described herein. In some embodiments, the subject is naive to treatment for MS. In certain embodiments, the subject is negative for hepatitis B virus (HBV). In some embodiments, the subject is negative for hepatitis B virus surface antigen (HBsAg). In some embodiments, the subject is negative for anti-hepatitis B virus core antibodies. In certain embodiments, the subject has not been immunized with a vaccine for at least 2 weeks or at least 4 weeks prior to treatment with the anti-CD20 antibody composition as described herein.
[00357] In some embodiments of the method, the subject is pre-medicated with an amount of a corticosteroid about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein. In some embodiments, the corticosteroid is methylprednisone or a corticosteroid bioequivalent thereto. In some embodiments, the amount of a corticosteroid is about 100 mg methylprednisone. In some embodiments, the corticosteroid is dexamethasone or a corticosteroid bioequivalent thereto. In some embodiments, the amount of a corticosteroid is from about 10 to 20 mg dexamethasone. In certain embodiments, the subject is pre-medicated with an amount of an antipyretic about 30 to about 60 minutes prior to administration of the anti- CD20 antibody composition as described herein. In some embodiments, the antipyretic is acetaminophen or an antipyretic bioequivalent thereto. In some embodiments, the subject is premedicated with an amout of an antihistamine about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein. In some embodiments, the antihistamine is diphenhydramine HC1 or an antihistamine bioequivalent thereof. In some embodiments, the amount of an antihistamine is from about 25 to 50 mg diphenhydramine HC1. In some embodiments, the subject is pre-medicated with an amount of a corticosteroid (as described above) and an amount of an antihistamine (as described above) about 30 to about 60 minutes prior to administration of the anti-CD20 antibody composition as described herein. In some embodiments, the corticosteroid and/or antihistamine are administered to the subject orally.
[00358] In some embodiments, the method alleviates or delays progression of one or more symptoms of MS in the subject. In certain embodiments, the method reduces the annualized relapse rate (ARR) in the subject following administration of the anti-CD20 antibody (e.g., ublituximab) composition as described herein. In some embodiments, ARR is the total number of relapse for a subject divided by the sum of treatment duration (i.e., it is the ratio of the sum of the subject’s RMS relapse counts divided by the sum of the subject’s treatment duration (in years)). In some embodiments, ARR is number of Independent Relapse Adjudication Panel (IRAP)- confirmed relapses for a subject per year. In some embodiments, reduction of ARR is assessed at about 96 weeks after the first infusion. In some embodiments, the subject achieves a significantly reduced ARR following administration of the multi-infusion dosage regimen of the anti-CD20 antibody composition described here, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. For example, as compared to teriflunomide administration, administration of the anti-CD20 antibody composition described here can significantly reduce ARR with a relative reduction in relapse rate of 59%.
Additionally, or in the alternative, as compared to teriflunomide administration, administration of the anti-CD20 antibody composition described here can significantly reduce ARR with a relative reduction in relapse rate of 49%. In some embodiments, treatment with the anti-CD20 antibody composition described here results in an ARR of about 0.100 to about 0.050; such as, about 0.100 to about 0.090, about 0.090 to about 0.080, about 0.080 to about 0.070, about 0.070 to about 0.060, or about 0.060 to about 0.050 (e.g., about 0.099, 0.098, 0.097, 0.096, 0.095, 0.094,
0.093, 0.092, 0.091, 0.090, 0.089, 0.088, 0.087, 0.086, 0.085, 0.084, 0.083, 0.082, 0.081, 0.080,
0.079, 0.078, 0.077, 0.076, 0.075, 0.074, 0.073, 0.072, 0.071, 0.070, 0.069, 0.068, 0.067, 0.066,
0.065, 0.064, 0.063, 0.062, 0.061, 0.060, 0.059, 0.058, 0.057, 0.056, 0.055, 0.054, 0.053, 0.052,
0.051, or 0.050). [00359] In some embodiments, the method reduces the total number of T1 gadolinium (Gd)- enhancing lesions in the subject following administration of the anti-CD20 antibody composition as described herein. In some embodiments, reduction of Gd-enhancing T1 lesions is assessed by MRI scan. In some embodiments, reduction of Gd-enhancing T1 lesions is assessed at about 96 weeks after the first infusion. In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody achieves a reduced total number of Gd-enhancing T1 lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, significantly reduced the total number of gadolinium-enhancing T1 lesions, with a relative reduction of about 97%.
[00360] In some embodiments, the method reduces the number of new or enlarging T2 hyperintense lesions in the subject following administration of the anti-CD20 antibody composition as described herein. In some embodiments, reduction of the number of new or enlarging T2 hyperintense lesions is assessed by MRI scan. In some embodiments, reduction of the number of new or enlarging T2 hyperintense lesions is assessed at about 96 weeks after the first infusion. In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves a reduced total number of new and enlarging T2 hyperintense lesions per MRI scan, as compared to a subject that receive orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, reduced the total number of new or enlarging T2 hyperintense lesions, with a relative reduction of about 90-92%.
[00361] In some embodiments, the method achieves confirmed disability progression in the subject following administration of the anti-CD20 antibody composition as described herein. In some embodiments, confirmation of disability progression comprises an increase of greater than or equal to 1.0 point from a baseline EDSS score of the subject attributable to MS, wherein said baseline EDSS score is 5.5 or less. In some embodiments, confirmation of disability progression comprises an increase of greater than or equal to 0.5 point from a baseline EDSS score of the subject attributable to MS, wherein said baseline EDSS score is greater than 5.5. [00362] In some embodiments, the method results in no evidence of disease activity (NED A) in the subject. In certain instances, NED A comprises one or more of no confirmed relapses, no gadolinium-enhancing (Gd+) T1 lesions, no new and/or enlarging T2 lesions, and no 12-week confirmed disability progression. In some embodiments, methods of the present disclosure results in NED A in the subject about 24 weeks after administration of a pharmaceutical formulation (e.g., an anti-CD20 antibody composition) as described herein. For example, administration of a pharmaceutical formulation described herein can result in NEDA in the subject about 24-96 weeks (e.g., 24-48 weeks, 24-72 weeks, 48-72 weeks, 72-96 weeks, or 48-96 weeks (e.g., 24 weeks, 36 weeks, 48 weeks, 60 weeks, 72 weeks, 84 weeks, 96 weeks, or any range in between)) after the administration. In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves increased NEDA status compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, increases NEDA status by 197%. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, increases NEDA status by 277%.
[00363] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves increased Confirmed Disability Improvement (CDI), as compared to a subjects that received orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide, increased CDI by 116% at 12 weeks. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, increased CDI by 103% at 24 weeks. [00364] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves an increased Multiple Sclerosis Functional Composite (MSFC) score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, administration of the anti-CD20 antibody (e.g., ublituximab), as compared to teriflunomide administration, increased the MSFC score by about 76%- 90%.
[00365] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves an improved timed 25-Foot Walk (T25FW) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period. In some embodiments, the subject administered the multiinfusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves an improved 9- Hole Peg test (9-HPT) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[00366] In some embodiments, the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody (e.g., ublituximab) achieves a significant reduction in both volume and number of new T1 hypointense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
[00367] In some embodiments, the treatment for MS (e.g., RMS), as disclosed herein, further comprises administering the anti-CD20 antibody in combination with one or more additional therapeutic agents. For example, the anti-CD20 antibody used in the present methods of treating RMS, can be given in combination with other compounds, drugs, and/or agents useful for the treatment of RMS. Such compounds, drugs, and/or agents can include, for example, small molecule drugs, monoclonal antibodies, or other B-cell depleting agents. In some embodiments, the methods described herein are used in combination with current standard of care MS treatment. In some instances, the anti-CD20 antibody is used in combination with a Bruton’s tyrosine kinase (BTK) inhibitor for treating RMS, including, e.g., fenebrutinib, evobrutinib, tolebrutinib, orelabrutinib BIIB091, AC0058, PRN473 (Dolgin, Nature Biotechnology 39:3-12 (2021), as well as those compounds described in U.S. Patent No. 9,951,077. In some instances, the anti-CD20 antibody is used in combination regimens that take advantage of non-overlapping mechanisms from ublituximab for B-cell depletion.
[00368] The anti-CD20 antibody (e.g., ublituximab) can be combined with injectable medications, oral medications, or infused medications. Injectable medications for combination therapy with the anti-CD20 antibody can include interferons, including but not limited to AVONEX (interferon beta-la), BETASERON (interferon beta-lb), EXTAYIA (interferon betalb), PLEGRIDY (peginterferon beta- la), and REBIF (interferon beta- la). Other injectable medications can include COPAXONE (glatiramer acetate). Orally available medications for combination with ublituximab can inlucde AUBAGIO (teriflunomide), fumarate based compositions including BAFIERTAM, dimethyl fumarate, TECFIDERA, and VUMERITY. Additionally, or in the altyernative, the anti-CD20 antibody (e.g., ublituximab) can be used in combination with the monoclonal antibody Tysabri (natalizumab). Orally administered agents that can be used in combination with the anti-CD20 antibody include GILENYI (fingolimod), MAYZENT (Siponimod), ZEPOSIA (ozanimod), and PONVORY (ponesimod). Additional orally administered agents include MAVENCLAD (cladribine).
[00369] The anti-CD20 antibody (e.g., ublituximab) can also be used in combination with infused medications, including, LEMTRADE (alemtuzumab) and NOVANTRONE (mitoxantrone).
[00370] In some embodiments, in subjects (e.g., patients) with a known resistance to one or more anti-CD20 antibodies, an anti-CD20 antibody of the present disclosure (e.g., ublituximab or an anti-CD20 antiody that binds the same epitope as ublituximab) can be administered, as ublituximab targets a unique epitope region on the CD20 antigen that is not targeted by other anti-CD20 monoclonal antibodies.
[00371] In some embodiments, additional therapeutic agents can comprises a B-cell depleting agent that is not an anti-CD20 antibody. In some instances, the B-cell depleting agent is a PI3K inhibitor.
[00372] In some embodiments, the additional therapeutic agent comprises a B-cell depleting agent that, like the anti-CD20 antibody (e.g., ublituximab) described here, is an anti-CD20 antibody. For example, ublituximab can be used in combination with additional anti-CD20 antibody compositions, such as, e.g., OCREVUS® (ocrelizumab), KESIMPTA® (ofatumumab), and Rituxan (rituximab).
[00373] The additional therapeutic agent can be administered orally, parenterally, intravenously, or subcutaneously.
[00374] In some embodiments, the method described herein can result in treatment-emergent adverse events (TEAEs). In some instances, TEAE comprises cytopenia or reduction in blood cell count in the subject. Blood cell count can be evaluated by blood tests, such as a complete blood count (CBC). Blood cell count can be obtained by cell counting methods known in the art, including, but not limited to manual methods (e.g., by using a hemocytometer) and automated methods (e.g., by using an automated cell counter). In some instances, cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60%, about 50-70%, about 60-70%, about 60-80%, about 70-80%, about 70-90%, about 80-90%, about 80-100%, about 90-100%, or any range in between (e.g., about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, about 100%, or any range in between)) reduction or decrease in blood cell count in a subject, as compared to a control or normal blood cell count. A control or normal blood cell count may comprise the blood cell count of a healthy subject who has not been administered the pharmaceutical formulation of the present disclosure. Additionally, or in the alternative, blood cell count of a subject prior to treatment by the present methods may serve as the control or normal blood cell count. Cytopenia or reduction in blood cell count can comprise one or more of lymphopenia, neutropenia, leukopenia, and anemia.
[00375] In some instances, administration of a pharmaceutical formulation (e.g., an anti-CD20 antibody composition), as described herein, results in cytopenia in the subject about 1-3 days (e.g., about 1 day, about 2 days, or about 3 days) after the administration. For example, administration of the pharmaceutical formulation can result in lymphopenia in the subject about 2 days after the administration. In certain instances, the cytopenia (e.g., lymphopenia, neutropenia, leukopenia, and/or anemia) is transient, such that blood cell count in the subject is normalized (e.g., becomes same or similar to control or normal blood cell count) by about 15 days (e.g., by 14 days, by 13 days, by 12 days, by 11 days, by 10 days, by 9 days, by 8 days, by 7 days, by 6 days, by 5 days, or by 4 days) after administration of the pharmaceutical formulation. For example, lymphocyte count in the subject can be normalized in the subject by 8 days after administration of the pharmaceutical formulation.
Methods of Treating Cancer
[00376] Also provided herein are methods of treating a cancer in a subject in need thereof, comprising administering to the subject an effective amount of an anti-CD20 antibody composition as described herein. In some embodiments, the cancer is a bladder cancer, a breast cancer, a cervical cancer, a colorectal cancer, a gynecologic cancer (i.e., cervical, ovarian, uterine, vaginal, or vulvar cancer), head and neck cancer, kidney cancer, liver cancer, lung cancer, lymphoma, mesothelioma, myeloma, prostate cancer, skin cancer, or thyroid cancer.
[00377] Also provided herein are methods of treating a disease or disorder associated with excessive B-cell proliferation in a subject in need thereof, comprising administering to the subject an effective amount of an anti-CD20 antibody composition as described herein. In some embodiments, the disease or disorder associated with excessive B-cell proliferation is a hematological cancer. In some embodiments, the hematological cancer is lymphoma, leukemia, or myeloma. In some embodiments, the hematological cancer is selected from B-cell lymphoma, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma (FL), Waldenstrom's macroglobulinemia (WM), diffuse large B-cell lymphoma (DLBCL), marginal zone lymphoma (MZL), hairy cell leukemia (HCL), Burkitt's lymphoma (BL), Richter's transformation, or primary central nervous system lymphoma (PCNSL). In various aspects of the method, the anti- CD20 antibody composition as described herein comprises anti-CD20 antibodies, which are IgGl antibodies comprising heavy chains each comprising the amino acid sequence of SEQ ID NO:1, and light chains each comprising the amino acid sequence of SEQ ID NO:2.
[00378] In some embodiments, the hematological cancer is B-cell lymphoma. In some embodiments, the B-cell lymphoma is relapsed or refractory. In certain embodiments, the hematological cancer is non-Hodgkin's lymphoma (NHL). In some embodiments, the NHL is relapsed or refractory. In certain embodiments, the hematological cancer is Waldenstrom's macroglobulinemia (WM). In some embodiments, the WM is relapsed or refractory. In certain embodiments, the hematological cancer is marginal zone lymphoma (MZL). In some embodiments, the MZL is relapsed or refractory. In certain embodiments, the hematological cancer is chronic lymphocytic leukemia (CLL). In some embodiments, the CLL is relapsed or refractory. In certain embodiments, the hematological cancer is small lymphocytic lymphoma (SLL). In some embodiments, the SLL is relapsed or refractory. In certain embodiments, the hematological cancer is primary central nervous system lymphoma (PCNSL). In some embodiments, the PCNSL is relapsed or refractory.
[00379] In some embodiments, the method of treating a hematological cancer comprises administering to the subject the anti-CD20 antibody composition as described herein by intravenous infusion. In some embodiments, the intravenous infusion comprises: a) an infusion comprising about 900 mg of the anti-CD20 antibody composition as described herein on days 1 and 2 (split into a 150 mg dose on day 1 and a 750 mg dose on day 2), day 8, and day 15 of Cycle 1 (each Cycle being 28 days); day 1 of Cycles 2-6; and day 1 of every 3 cycles after Cycle 6 (e.g., Cycle 9, 12, 15, etc.). In some embodiments, the hematological cancer is CLL. [00380] In some embodiments, the method described herein can result in treatment-emergent adverse events (TEAEs). In some instances, TEAE comprises cytopenia or reduction in blood cell count in the subject. Blood cell count can be evaluated by blood tests, such as a complete blood count (CBC). Blood cell count can be obtained by cell counting methods known in the art, including, but not limited to manual methods (e.g., by using a hemocytometer) and automated methods (e.g., by using an automated cell counter). In some instances, cytopenia comprises about 20-100% (e.g., about 20-30%, about 20-40%, about 30-40%, about 30-50%, about 40- 50%, about 40-60%, about 50-60%, about 50-70%, about 60-70%, about 60-80%, about 70-80%, about 70-90%, about 80-90%, about 80-100%, about 90-100%, or any range in between (e.g., about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, about 100%, or any range in between)) reduction or decrease in blood cell count in a subject, as compared to a control or normal blood cell count. A control or normal blood cell count may comprise the blood cell count of a healthy subject who has not been administered the pharmaceutical formulation of the present disclosure. Additionally, or in the alternative, blood cell count of a subject prior to treatment by the present methods may serve as the control or normal blood cell count. Cytopenia or reduction in blood cell count can comprise one or more of lymphopenia, neutropenia, leukopenia, and anemia.
Multi-infusion Dosage
[00381] In any of the embodiments described herein, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1; b) a second infusion comprising about 500 to about 700 mg, about 800 to about 1000 mg, or about 1100 to about 1300 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 500 to about 700 mg, about 800 to about 1000 mg, or about 1100 to about 1300 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; d) a fourth infusion comprising about 500 to about 700 mg, about 800 to about 1000 mg, or about 1100 to about 1300 mg of the anti-CD20 antibody composition at about 3 weeks after the first infusion; and e) one or more subsequent infusions comprising about 400 to about 500 mg, about 500 to about 700 mg, about 800 to about 1000 mg, or about 1100 to about 1300 mg of the anti-CD20 antibody composition at about one month after the prior infusion. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; the fourth infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
[00382] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosy lated glycans) at day 1 ; b) a second infusion comprising 600, 900, or 1200 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising 600, 900, or 1200 mg of the anti- CD20 antibody composition at about 2 weeks after the first infusion; d) a fourth infusion comprising 600, 900, or 1200 mg of the anti-CD20 antibody composition at about 3 weeks after the first infusion; and e) one or more subsequent infusions comprising 450, 600, 900, or 1200 mg of the anti-CD20 antibody composition at about one month after the prior infusion. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; the fourth infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
[00383] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 400 to about 500 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 500 to about 700 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 800 to about 1000 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusions comprising about 400 to about 1000 mg of the anti- CD20 antibody composition at about one month after the prior infusion. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
[00384] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 450 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising 600 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising 900 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusions comprising 450, 600, or 900 mg of the anti-CD20 antibody composition at about one month after the prior infusion. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
[00385] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 5 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; and b) one or more subsequent infusions comprising about 5 to about 450 mg of the anti-CD20 antibody composition at about one week after the prior infusion, wherein each subsequent infusion is at higher dose than the prior infusion. In some instances, the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over 1 hour.
[00386] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; and b) one or more subsequent infusions comprising about 400 to about 500 mg of the anti-CD20 antibody composition at about one week after the prior infusion. In some instances, the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over 1 hour.
[00387] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody composition at day 1; and b) one or more subsequent infusions comprising 450 mg of the anti- CD20 antibody composition at about one week after the prior infusion. In some embodiments, the method comprises seven or more subsequent infusions. In some embodiments, the method comprises seven subsequent infusions. In some instances, the first infusion is administered over 4 hours; and the one or more subsequent infusions are administered over 1 hour.
[00388] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody at about 1 week after the first infusion; c) a third infusion comprising about 500 to about 1000 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusions comprising about 500 to about 1000 mg of the anti-CD20 antibody composition at about one month after the prior infusion. In certain embodiments, the one or more subsequent infusions each comprise 600 mg of the anti-CD20 antibody composition as described herein. In certain embodiments, the first infusion, second infusion, third infusion, and one or more subsequent infusions each comprise 600 mg of the anti-CD20 antibody composition as described herein. In some embodiments, the first infusion, second infusion, third infusion, and one or more subsequent infusions each comprise 900 mg of the anti-CD20 antibody composition as described herein. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour.
[00389] In some embodiments, the intravenous infusion comprises a multi-infusion dosage regimen, comprising: a) a first infusion comprising about 100 to about 200 mg of the anti-CD20 antibody composition as described herein (i.e., the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 10 to 20% galactosylated glycans and about 20 to 40% fucosylated glycans) at day 1 ; b) a second infusion comprising about 700 to about 800 mg of the anti-CD20 antibody composition at about 1 week after the first infusion; c) a third infusion comprising about 850 to about 950 mg of the anti-CD20 antibody composition at about 2 weeks after the first infusion; and d) one or more subsequent infusions comprising about 850 to about 950 mg of the anti-CD20 antibody composition at about one month after the prior infusion. In some embodiments, the first infusion comprises 150 mg of the anti-CD20 antibody composition, the second infusion comprises 750 mg of the anti-CD20 antibody composition, the third and one or more subsequent infusions comprise 900 mg of the anti-CD20 antibody composition. In some instances, the first infusion is administered over 4 hours; the second infusion is administered over 1 hour; the third infusion is administered over 1 hour; and the one or more subsequent infusions are administered over 1 hour. Subject
[00390] In some embodiments, the subject is a human. In some embodiments, the human subject is an adult. In some embodiments, the subject has relapsed or refractory B-cell lymphoma. In some embodiments, the subject has been previously treated with at least one prior course of rituximab or a rituximab-based therapy. In some embodiments, the subject has been previously treated with at least one prior course of fludarabine or a fludarabine-based therapy. In some embodiments, the subject is naive to treatment for a B-cell lymphoma. In some embodiments, the subject is eligible for high dose or combination chemotherapy and/or stem cell transplant. In some embodiments, the subject has an Eastern Cooperative Oncology Group (ECOG) score of from 0 to 2 prior to treatment with the anti-CD20 antibody composition as described herein. In some embodiments, the subject has a peripheral blood lymphocyte count of greater than 5,000/pL prior to treatment with the anti-CD20 antibody composition as described herein.
[00391] In some embodiments, the anti-CD20 antibody compositions provided herein can be used to treat and/or prevent Chronic inflammatory demyelinating polyneuropathy (CIDP); Myositis; Lupus Nephritis; other forms of MS- PPMS, SPMS; Myasthenia Gravis (MG); Antiphospholipid Syndrome; Thrombotic Thrombocytopenic Purpura (TTP); Ulcerative Colitis; Minimal Change Nephrotic Syndrome (MCNS); Aplastic Anemia; Autoimmune Glomerulopathies; Rheumatoid Arthritis (RA); Interstitial Lung Disease; Myasthenia Gravis (MG); Subepidermal Autoimmune Blistering Disease; Pulmonary Infectious Diseases; Acquired Hemophilia; Refractory Mixed Cryoglobulinemia; Primary Immune Thrombocytopenia; Graft Versus Host Disease (GVHD); Autoimmune Blistering Disease; Anti-Myelin Associated Glycoprotein (MAG) Polyneuropathy; Granulomatosis with Pollyangiitis (GPA); Neuromyelitis Optica; Systemic Lupus Erythematosus; Pemphigus; Post-Transplant Lymphoproliferative Disorders; Autoimmune Hemolytic Anemia; Cerebral Vasculitis; Microscopic Polyangiitis (MPA); or Idiopathic Nephritic Syndrome.
7.9 Pharmacokinetic Properties
[00392] Also provided herein are methods of treating a human patient afflicted with a disease (e.g., an autoimmune disease) comprising administering to the patient the anti-CD20 antibody provided herein. [00393] In some embodiments, the anti-CD20 antibody are administered as i) a first infusion at a dose of about 150 mg, ii) a second infusion two week later at a dose of about 450 mg, and iii) subsequent infusions every six months at a dose of about 450 mg.
[00394] In some embodiments, the administration of the anti-CD20 antibody produces an area under the curve (AUC) of between about 2,160 pg/mL and about 3,840 pg/mL. In certain embodiments, the AUC is about 3,000 pg/mL. In certain embodiments, the AUC is the steady state AUC.
[00395] In some embodiments, the administration of the anti-CD20 antibody produces a Cmax of between about 118,011 ng/mL and about 159,989 ng/mL. In certain embodiments, the Cmax is about 139,000 ng/mL. In certain embodiments, the Cmax is the steady state Cmax.
[00396] In some embodiments, the administration of the anti-CD20 antibody produces a Cmin of about 0 ng/mL and about 375 ng/mL. In certain embodiments, the Cmin is about 139 ng/mL. In certain embodiments, the Cmin is the steady state Cmin.
[00397] In some embodiments, the administration of the anti-CD20 antibody produces a Cavg of between about 6,437 ng/mL and about 11,443 ng/mL. In certain embodiments, the Cavg is about 8,940 ng/mL. In certain embodiments, the Cavg is the steady state Cavg.
[00398] In some embodiments, the autoimmune disease is selected from the group consisting of multiple sclerosis, psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, osteoporosis, eczema, allogenic or xenogenic transplantation (organ, bone marrow, stem cells and other cells and tissues), graft rejection, graft-versus-host disease, lupus erythematosus, inflammatory disease, type 1 diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis (e.g., Hasimoto's and autoimmune thyroiditis), myasthenia gravis, autoimmune hemolytic anemia, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic conjunctivitis, atopic dermatitis, chronic obstructive pulmonary disease, glomerulonephritis, neuroinflammatory diseases, and uveitis.
[00399] In some embodiments, the autoimmune disease is multiple sclerosis. In certain embodiments, the multiple sclerosis is a relapsing form of multiple sclerosis. In certain embodiments, the relapsing form of multiple sclerosis is a clinically isolated syndrome (CIS); relapsing-remitting MS (RRMS); active secondary progressive MS (SPMS); or primary progressive MS (PPMS). In certain embodiments, the relapsing form of MS is a clinically isolated syndrome (CIS). In certain embodiments, the relapsing form of MS is relapse-remitting multiple sclerosis (RRMS). In certain embodiments, the relapsing form of MS is active secondary progressive multiple sclerosis (SPMS). In certain embodiments, the relapsing form of MS is primary progressive MS (PPMS).
[00400] In certain embodiments, the anti-CD20 antibody is administered intravenously.
7.10 Methods of Making
[00401] Also povided herein are methods of making the populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications provided herein. Example 15 provides exemplary methods of making the populations of anti-CD20 antibody proteins provided herein.
[00402] In some embodiments, the methods of making the populations of anti-CD20 antibody proteins with specified ranges of post-translational modifications as described above comprises: i) culturing the rat hybridoma cells at a first culture pH of about 7.0 to about 7.55 for 0 to 3 days, ii) culturing the rat hybridoma cells at a second culture pH of about 6.5 to about 6.99 on day 3, iii) maintaining the culture pH at the second culture pH of about 6.5 to about 6.99 from culture day 3 until day 14 of the cell culture, and iv) controlling culture pCCh levels to less than about 200 mmHg throughout the culture period. In some embodiments, the second culture pH is about 6.60 to about 6.96 (e.g., the second culture pH is 6.8).
[00403] In some embodiments, the second culture pH results in higher integrated viable cell density (IVCD) and higher titer at harvest.
[00404] In some embodiments, the second culture pH results in lower percent fucosylation.
[00405] In some embodiments, the rat hybridoma cells expressing the recombinant protein are cultured in a basal media that is chemically defined and animal-derived component free (ADCF) culture medium
[00406] In some embodiments, the basal medium is supplemented with a feed medium.
[00407] In some embodiments, the method further comprises an initial temperature set point of about 37°C, wherein said initial temperature set point is set on culture day 0 to culture day 1.
[00408] In some aspects, the method further comprises a second temperature set point of about 35°C, wherein said second temperature set point is set at the end of culture day 1 to culture day 3. [00409] In some embodiments, the method further comprises comprising a third temperature set point of about 32°C to about 33°C, wherein said third temperature set point is set on culture day 3 and is maintained though harvest. The term “harvest” refers to the point of time in the mammalian cell culture process when cells containing the recombinant protein are separated and removed from the cell culture media and subject to additional processing, such as, e.g., centrifugation, filtration, or purification.
[00410] In some embodiments, harvest of the cells will occur on process day 12, 13, or 14 of the cell culture, or when cell viability drops below 20%, whichever comes first.
[00411] In some embodiments, the method further comprises harvesting the recombinant protein produced by the rat hybridoma cell.
[00412] In some embodiments, the method further comprises purifying the recombinant protein by affinity chromatography and/or ion exchange chromatography. In some embodiments, the affinity chromatography comprises protein A purification.
[00413] In some embodiments, the methods result in an increased yield of recombinant protein. For example, the recombinant protein is increased by at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 110 %, at least about 120%, at least about 130%, at least about 140%, or at least about 150%, relative to a recombinant protein produced by a culturing process that does not employ the culture conditions as recited above.
Sequence Table
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Ill
Figure imgf000114_0001
Figure imgf000115_0001
7.11 Preparing Ublituximab for Intravenous Infusion in the Disclosed Methods of Treatment
[00414] In some embodiments, the ublituximab infusions are prepared in 250 mb of 0.9% Sodium Chloride Injection. For example, ublituximab infusions can be prepared in infusion bags containing 0.9% Sodium Chloride Injection.
[00415] In some embodiments, the first infusion of 150 mg ublituximab is prepared by: (1) inspecting one vial of 150 mg/6 mL of ublituximab solution for any particulate matter or discoloration, and not using the solution if the solution contains discrete foreign particulate matter; (2) if no particulate matter in said vial, preparing 250 mL infusion bag for first infusion by using one vial of 150 mg/6 mL ublituximab solution; (3) withdrawing 6 mb 0.9% Sodium Injection from the infusion bag, and discarding; (4) withdrawing 6 mL ublituximab from said vial; (5) diluting 6 mL (150 mg) of ublituximab into said 250 mL infusion bag containing 0.9% Sodium Injection for immediate administration; and (6) mixing said infusion bag by gentle inversion, without shaking.
[00416] In some embodiments, the second or subsequent infusions of 450 mg ublituximab are prepared by: (1) inspecting three vials of 150 mg/6 mL of ublituximab solution for any particulate matter or discoloration, and not using the solution if the solution contains discrete foreign particulate matter; (2) if no particulate matter in said vial, preparing 250 mL infusion bag for second or subsequent infusion by using three vials of 150 mg/6 mL ublituximab solution; (3) withdrawing 18 mL 0.9% Sodium Injection from the infusion bag, and discarding; (4) withdrawing 18 mL ublituximab from said vial; (5) diluting 18 mL (450 mg) of ublituximab into said 250 mL infusion bag containing 0.9% Sodium Injection for immediate administration; and (6) mixing said infusion bag by gentle inversion, without shaking.
[00417] In some embodiments, prior to the start of ublituximab intravenous infusion, the content of the infusion bag is at room temperature.
7.12 Administration of Ublituximab in the Methods of Treatment Dislosed Herein
[00418] In some embodiments, prior to intitiating ublituximab administration, the subject is pre-screened for Hepatitis B virus (HBV). If the subject tests positive for Hepatitis B surface antigen (HbsAg), ublituximab should not be administered. In some embodiments, the subject being administered the ublituximab infusion is hepatitis B virus surface antigen (HBsAg) negative.
[00419] In some embodiments, the subject is pre-medicated with a corticosteroid and an antihistamine, 30-60 minutes prior to administration of ublituximab or an anti-CD20 antibody that binds the same epitope as ublituximab, both of which may be administered orally or intravenously. In some embodiments, the corticosteroid and/or antihistamine are administered to the subject orally. In some instances, the pre-treatment dosage of corticosteroid is about 100 mg methylprednisone, 10-20 mg dexamethasone, or an equivalent corticosteroid. [00420] In some embodiments, ublituximab is administered in a multi-dosage regimen. In certain instances, ublituximab is administered by intravenous infusion. For example, ublituximab can be administered in a multi-infusion dosage regimen by intravenous infusion. [00421] In some embodiments, ublituximab is administered to a patient with RMS by intravenous infusion in a multi-infusion dosage regimen for 48 weeks. In some embodiments, ublituximab is administered to a patient with RMS by intravenous infusion in a multi-infusion dosage regimen for 96 weeks.
[00422] The multi-infusion dosage regimen can comprise first, second, and subsequent intravenous infusions of ublituximab. In some embodiments, “subsequent infusions” of ublituximab can be any number of infusions after the first and second infusion.
[00423] In some embodiments, the multi-infusion dosage regimen comprises first and second infusions of 150 mg ublituximab intravenous infusion (First Infusion), followed two weeks later by a 450 mg ublituximab intravenous infusion (Second Infusion). In some embodiments, the multi-infusion dosage regimen further comprises subsequent infusions of 450 mg ublituximab intravenous infusion every 6 months.
[00424] In some embodiments, the duration of the first infusion of ublituximab, or an anti- CD20 antibody that binds the same epitope as ublituximab, is about four hours. For example, the infusion rate of the first infusion of ublituximab, or an anti-CD20 antibody that binds the same epitope as ublituximab, can be 10 mb per hour for the first 30 minutes; 20 mb per hour for the next 30 minutes; 35 mb per hour for the next hour; and 100 mb per hour for the remaining two hours. In some embodiments, the duration of the second and subsequent infusions of ublituximab is about one hour. For example, the infusion rate of the second and/or subsequent infusion of ublituximab can be 100 mb per hour for the first 30 minutes, and 400 mb for the remaining 30 minutes. The infusion duration may take longer if infusion is interrupted or slowed.
[00425] In some embodiments, the subject is monitored for at least one hour after the completion of the first two ublituximab infusions. Subsequent infusions do not require monitoring post-infusion unless an infusion-related reaction (IRR) and/or hypersensitivity has been observed.
[00426] In some embodiments, the ublituximab intravenous infusion is stopped and permanently discontinued if the subject has symptoms of a life-threatening infusion-related reaction. In some embodiments, the ublituximab intravenous infusion is discontinued if the subject has symptoms of a severe infusion-related reaction, and restarted once the subject’s infusion-related reaction symptoms have resolved. In some embodiments, the ublituximab intravenous infusion is restarted at half the infusion rate at the time of onset of the infusion- related reaction. The ublituximab infusion rate can be increased to the original infusion rate if the subject tolerates the half infusion rate. In some embodiments, the ublituximab infusion rate is reduced in half if the subject has symptoms of a mild to moderate infusion-related reaction, wherein the half infusion rate is maintained for at least 30 minutes. The ublituximab infusion rate can be increased to the original infusion rate if the subject tolerates the half infusion rate. [00427] In some embodiments, the administration of ublituximab by intravenous infusion results in a geometric mean steady-state AUC of 3000 mcg/mL per day (CV = 28%) and a mean maximum concentration of 139 mcg/mL (CV =15%).
[00428] In some embodiments, the administration of ublituximab can be via routes of admistration other that intravenous infusion (e.g., subcutaneous injection, intramuscular injection, oral, epidermal, spinal, or inhalation).
7.13 Pharmaceutical Compositions
[00429] " Pharmaceutical composition" can refer to a composition that is acceptable for pharmaceutical administration, such as to a human being. Such a composition can include substances that are impurities at a level not exceeding an acceptable level for pharmaceutical administration (such level including an absence of such impurities), and can include pharmaceutically acceptable excipients, vehicles, carriers, stabilizers, and other inactive ingredients, for example, to formulate such composition for ease of administration, in addition to any active agent(s).
[00430] The present disclosure provides pharmaceutical compositions comprising ublituximab, or an antibody that binds the same epitope as ublituximab, for use in the treatment of subjects with relapsing forms of Multiple Sclerosis, according to any of the methods disclosed herein.
[00431] In some embodiments, the pharmaceutical composition comprises ublituximab. In some embodiments, the pharmaceutical composition comprising ublituximab is formulated together with a pharmaceutical carrier. Suitable pharmaceutical carriers are known to those skilled in the art (Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA (1990)).
[00432] The pharmaceutical composition can comprise any number of excipients. Excipients that can be used include carriers, surface active agents, thickening or emulsifying agents, solid binders, dispersion or suspension aids, solubilizers, colorants, flavoring agents, coatings, disintegrating agents, lubricants, sweeteners, preservatives, isotonic agents, and combinations thereof. The selection and use of suitable excipients is taught in Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Ed. (Lippincott Williams & Wilkins 2003), the disclosure of which is incorporated herein by reference.
[00433] Pharmaceutical compositions described here can be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound can be coated in a material to protect it from the action of acids and other natural conditions that may inactivate it. As used herein, "parenteral administration" can mean modes of administration other than enteral and topical administration, usually by injection, and can include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Additionally, or in the alternative, pharmaceutical compositions described here can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
[00434] In some embodiments, the pharmaceutical composition comprising ublituximab is for administration to a subject by intravenous infusion.
[00435] The following examples are offered by way of illustration, and not by way of limitation.
8. EXAMPLES
8.1 Example 1 — Glycosylation Profile
[00436] The glycosylation profile of a sample of anti-CD20 antibody proteins provided herein was determined by measuring fluorescently labeled N-glycans (fluorescent label is 2- aminobenzamide) that were enzymatically cleaved from the anti-CD20 antibody proteins using PNGase F. The labeled glycans are resolved using a hydrophilic interaction column equipped. The glycans flow through a fluorescence detector after separation. Peak identification from the test sample chromatograms is identified based on retention time and relative to peaks in the glycan standard which have been confirmed by mass spectrometry. The relative percentage of each N-glycan is calculated based on the N-glycan peak area divided by the total peak area of all N-glycans. The glycosylation profile is shown in Figure 2.
8.2 Example 2 — Intact mass method
[00437] The glycosylation profile of anti-CD20 antibody proteins provided herein was assessed by intact mass analysis (LC-MS) under non-reducing conditions. The sample of anti- CD20 antibody proteins provided herein is first exchanged into MS appropriate buffers during the chromatography step using SEC and a mobile phase containing TFA, acetonitrile, and water. The sample is then introduced into an ESI-QTOF for intact mass analysis. The mass spectra are deconvoluted and the peaks are assigned based on mass. The relative abundance of each anti- CD20 antibody proteins provided herein containing N-glycan was calculated by taking the abundance of an N-glycan and dividing by the total abundance of all identified peaks. Results are provided in the table below and in Figure 3.
Table 8: Intact Molecular Weights for Sample of anti-CD20 antibody proteins by LC-MS
Figure imgf000120_0001
Figure imgf000121_0001
Abbreviation: ND = not detected. 8.3 Example 3 — Cell-Based ADCC Assay
(a) Materials and Method
[00438] Antibody dependent cytotoxicity (ADCC) is mediated through binding of the Fc portion of TG-1101 (TG Therapeutics, Inc.) to the FcylllA Receptor on the effector cells. The assay used for this analysis employs Eurofins- DiscoverX’s “KILR CD 16a effector cells”, which are single donor-derived human CD8+ T-lymphocytes engineered to express CD16 (FcyRIII) on their plasma membrane surface. These cytotoxic T cells provide reduced background killing, increased accuracy and reproducibility compared to PMBC preparations isolated from fresh blood. Raji cells are used as target cells, and ADCC activity is determined from the lysis of the target cells.
[00439] KILR cells are obtained from Eurofins, and Raji cells from ATCC. Master and Working cell bank system was used to ensure quality of the reagents. Raji cells were seeded at 1x105 cells/mL, KILR Effector cells were seeded at 5x105 cells/mL, and final effector: target (E:T) ratio was 5:1. An eight-point dilution series of the samples were used in the concentration range of 250.00 pg/ml - 0.04 pg/ml (250, 50, 16.7, 5.6, 1.9, 0.6, 0.2, 0.04 pg/ml). Cell mixtures and testing samples were cultured at 36±1°C, 5±1%CO2 for 18-22 hours. At the end of the incubation, a CytoTox GLo™ preparation is added, and plates are incubated for 30±10 minutes. The plates are read using the SpectraMax plate reader. Two independent preparations of the materials are prepared and assayed across duplicate plates. Assay controls are prepared in triplicates and include: Target Cells Alone Control, Target Cells Death Control, Effector Cells Alone Control, and Effector & Target Cells Control. SoftMax Pro was used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. Results are reported as percentage ADCC activity compared to Ublituximan reference standard. EC50 of each testing sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP482, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
(b) Results
[00440] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 4, all anti-CD20 samples exhibited dose- dependent ADCC activity, TG-1101 (TG Therapeutics, Inc.) has highest ADCC activity compared to other anti-CD20. The ADCC activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 9, compared to TG-1101 (TG Therapeutics, Inc.), Gazava has ADCC activity that is relative similar to TG-1101 (TG Therapeutics, Inc.), while the ADCC activities of Rituxan, Ocrevus, and Arzerra are significantly lower. The ED50s of each anti-CD20 are also shown in Table 9, overall TG-1101 (TG Therapeutics, Inc.) and Gazyva have lower EC50 than Arzerra, Rituxan, and Ocrevus. EC50 of TG-1101 (TG Therapeutics, Inc.) is approximately 25 fold lower than Ocrevus.
Table 9: ADCC Activity and EC50
Figure imgf000123_0001
8.4 Example 4 — Cell-Based ADCC Assay Using Primary NK Cells
(a) Materials and Method
[00441] This ADCC assay was performed using CD20 expressing Raji cells as target cells, primary NK cells as effector cells, and LDH as target cell lysis read-out. Raji cells (ATCC, Cat# CCL-86TM) were seeded atlxl05 cells/well on plates. Primary NK cells isolated from human donor PBMC using Miltenyibiotec kit (Cat# 130-092-657) . E/T ratio of 5: 1 for NK92/CD16a cells and primary NK cells was used in the assay. An eight-point dilution series of the samples, in triplicates, were used in the concentration range of 0.01 pg/ml - 0 ug/ml with a dilution factor of 10. The target cells were incubated with test sample dilutions for 30 min in 37°C incubator. Effector cells were added to the target cell cultures followed by 6 hours incubation after which supernatants were collected. The background (OD650nm) subtracted OD492nm data were used to calculate the LDH release. The percentages of cell lysis was calculated according the formula below: Cell lysis %=100*(ODSample data- ODtumor cells plus effector cells) / (ODMaximum release - OD Minimum release). (b) Results
[00442] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, and Arzerra. As shown in Figure 5, all anti-CD20 samples exhibited dose-dependent ADCC activity. The calculated EC50 values are summarized in Table 10. TG-1101 (TG Therapeutics, Inc.) and Gazyva displayed higher ADCC activities and lower EC50 than Rituxan and Arzerra.
Table 10: EC50 of ADCC assay Using Primary NK Cells
Figure imgf000124_0001
8.5 Example 5 — Cell-Based ADCP Assay
(a) Materials and Method
[00443] Antibody dependent cellular phagocytosis (ADCP) is another potential mechanisms of action (MO As) for anti-CD20s. ADCP activity was assessed using an assay in which CD20 expressing Daudi cells were used as target cells (ATCC, Cat# CCL-213, labeled by PKH26). Human monocytes were isolated from PBMC from 20 human donors (using human Pan Monocyte Isolation Kit, MiltenyiB iotec, Cat#130-096-537) and differentiated in vitro using GM- CSF to yield macrophages. An E/T ratio of 5:1 was used; and the ADCP was assessed by flow cytometry in this assay. An eight-point dilution series of the samples, in duplicates and in the concentration range of 100 pg/ml - 0 ug/ml with a dilution factor of 10, were incubated with PKH26-labeled target cells. Macrophages were then co-cultured with PKH26-labeled target cells for 22 hours. Target cell phagocytosis was assessed by flow cytometry. Controls in the assay included Target cell control of PKH26 stained Daudi cells only; Effector cell control of PKH67 stained MDM only.
[00444] Effector and target cells control with a non-specific IgGl antibody; Effector and target cells control (background control). ADCP was determined by FACS as a percentage of PKH26/PKH67 double positive cell counts/PKH26.
(b) Results
[00445] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, and Arzerra. As shown in Figure 6, all anti-CD20 samples exhibited dose-dependent ADCP activity. The calculated EC50 values are summarized in Table 11. The EC50s are in the ng/ml range, and given the nature of the test method, the ADCP activities of all samples are considered similar.
Table 11: EC50 of ADCP Assay
Figure imgf000125_0001
Blinding Code: 50K068:70T:2003=Gazyva; 52W243:70T:2003=ARZERRA;
54A157:70T:2003=Rituxan
8.6 Example 6 — Cell Based CDC Assay
(a) Materials and Method
[00446] The Complement Dependent Cytotoxicity (CDC) is mediated through binding of the Fc portion of TG-1101 (TG Therapeutics, Inc.) to Cl q Receptor in the complement system. The CDC activity assay used in this analysis is a cell-based assay using the CD20 expressing human mantle cell lymphoma cell line, Jeko-1 and rabbit serum as the source of complement. CDC mediated cell lysis is measured by the Cell Titer-Gio™ reagent (Promega). A nine-point dilution series of the samples are used in the concentration range of 10,000 ng/ml - 10.42 ng/ml (10,000.00, 3333.33, 1666.67, 833.33, 416.67, 208.33, 104.17, 52.08, 10.42 ng/ml). Two independent preparations of each sample are prepared and assayed across duplicate plates. Assay negative controls are prepared in triplicates and include target cells & complement control and target cells alone control.
[00447] Jeko-1 cells, obtained from the ATCC and maintained through a master banking system, were seeded at 3x105 cells/mL and incubated for 60-90 minutes. Samples dilutions, and then complement were added, and the plates were incubated for approximately 2 hours at 37°C and 25 minutes at room temperature. Target cells with complement only control and target cell only control provided a basal level of target cell viability over the course of the assay. The Cell Titer-Gio reagent is then added and incubated an additional 30 minutes at room temperature. At the end of the assay the plates are read using a SpectraMax M5 plate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. The resulting data is evaluated using the PLA software for parallelism and potency against the reference standard. Results are reported as % potency relative to the reference standard. EC50 of each testing sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP463, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
(b) Results
[00448] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 7, all samples exhibited dose-dependent CDC activity, except for Gazyva, which is known to have reduced CDC activity. Rituxan and Arzerra have comparable CDC activity; Ubli and Ocrevus have comparable CDC activity. The CDC activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 12, compared to TG-1101 (TG Therapeutics, Inc.), Gazava has ADCC activity that is relative similar to TG-1101 (TG Therapeutics, Inc.), while the ADCC activities of Rituxan, Ocrevus, and Arzerra are significantly lower. The ED50s of each anti-CD20 are also shown in Table 12, which shows similar comparisons as the level of CDC activities.
Table 12: CDC Activity and EC50
Figure imgf000126_0001
8.7 Example 7 — Cell Based CD20 Binding Assay
(a) Materials and Method
[00449] The CD20 binding used for this analysis is a cell based binding assay that uses a CD20 expressing human mantle cell lymphoma cell line, Jeko-1, and an MSD (MesoScale Discovery) assay format. Jeko-1 target cells are seeded onto MSD plates, test samples are incubated and allowed to bind to Jeko-1 cells, anti- human Fc detection antibody conjugated with streptavidin-SULFOTAG™ is used to emit electrochemiluminescence signal. An eight-point dilution series of test samples were used in the concentration range of 40,000.00 pg/ml - 0.23 ng/ml (40,000.00, 4,000.00, 1,000.00, 333.30, 111.10, 37.00, 4.60, 0.23 ng/ml). Two independent preparations of the Test Material are prepared for each 2-plate assessment. Assay controls include No cell control (Reference Standard/Test Material dilution + detection reagent, omitting cells) and Cell only control (Cells + detection reagent, omitting Reference Standard/Test Material).
[00450] Jeko-1 cells, obtained from the ATCC and managed through a master banking system, are seeded onto MSD high bind plate in PBS at 3 x 105 cells per mL, in a final volume of 100 pL per well, and incubated at 35-37°C for 2 hours ±10 min. Unbound cells are removed by a PBS wash, the plates are blocked then washed. Fifty pL of sample dilutions were added, and the plate is incubated at room temperature for 1 hour ±10 min while shaking. Following incubation and three washes, 50 pL anti-human Fc detection antibody conjugated with STREP-SULFOTAG is added and incubated for 1 hour ± 10 min at room temperature while shaking. The plates are washed again, and 150 pL of the MesoScale read buffer, containing tripropylamine (TP A), is added as a co-reactant for light generation for an electrochemiluminescence read out. Plates are read immediately on a MSD Reader using Workbench 4.0. The resulting data is evaluated using the PLA software and analyzed using a constrained 4 parameter logistic model. Binding activity results are reported as percentage potency relative to the reference standard. EC50 of each testing sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP466, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
(b) Results
[00451] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control. As shown in Figure 8, all samples exhibited dose-dependent CD20 binding. With the exception of Gazyva, which is a Type II anti-CD20 and is known to have approximately 50% of target occupancy, Ubituximab, Rituxan, Ocrevus, and Arzerra have similar maximum binding. The CD20 binding activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard and binding EC50s are shown in Table 13. The CD20 binding affinity of the 4 anti-CD20s are similar. Table 13: CD20 Binding Activity and EC50
Figure imgf000128_0001
[00452] EC50s values listed are negative, rising from the PLA software log transforms the concentrations to base 2. The actual ED50 (in pg/ml) is 2n, where n = the number listed.
8.8 Example 8 — Cell Surface CD20 Binding By FACS
(a) Materials and Method
[00453] The binding of TG-1101 (TG Therapeutics, Inc.) to cell surface CD20 on Raji and Daudi cells was characterized by FACS analysis at LakePharma. A six-point dilution series of the samples, in duplicates and in the concentration range of 40 pg/ml - 0 ug/ml with a dilution factor of 5 were used. Cells were incubated with sample dilutions; binding was detected using a PE conjugated anti-human secondary antibody.
(b) Results
[00454] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Arzerra, and Rituxan. As shown in Figure 9, all anti-CD20 samples exhibited dose-dependent CD20 binding to both Daudi and Raji cells. The binding characteristics as evaluated by the FACS assay are similar to that evaluated by the MSD assay. With the exception of Gazyva, which is a Type II anti-CD20 and is known to have approximately 50% of target occupancy, Ubituximab, Rituxan, and Arzerra have similar maximum binding. The calculated EC50 values are summarized in Table 14. The CD20 binding affinity of the four anti-CD20 antibodies are similar. Table 14: EC50 of Cell Surface CD20 Binding by FACS
Figure imgf000129_0001
8.9 Example 9 — FcyRIIIA Binding Assay
(a) Materials and Method
[00455] The assay used in this analysis is a surface plasmon resonance (SPR)-based method that measures the binding to both FcyRIIIa 158V and FcyRIIIa 158F receptors. The method follows a direct binding assay methodology where the FcyRIIIa receptor is directly immobilized onto the flow cell on a sensor chip surface and samples are injected over the chip to assess binding. FcyRIIIa 158V receptor (3pg/ml) or FcyRIIIa 158F receptor (6 pg/ml) is immobilized on the chip surface using covalent amine coupling chemistry. Eight-point dilution series of the testing samples are prepared in the concentration range of 1000 nM - 15.6 nM with a dilution factor of 2. Independent duplicates of sample dilutions are injected over the chip, followed by surface regeneration between each cycle. The binding is measured in response units (RU). The kinetics of the binding reaction is determined by measuring changes in SPR due to the increase in mass in the close proximity to the biosensor chip surface. Change in the mass of the complex as a function of time is visualized as a sensorgram.
[00456] The equilibrium dissociation constants (KD) of each sample is determined for each receptor. The rates of change of the SPR signal is analyzed using a 1 : 1 Langmuir model for FcyRIIIa 158V variant to yield apparent rate constants for the association and dissociation phases of the reaction, and equilibrium dissociation constants. KD is determined using steady state affinity for the FcyRIIIa 158F variant. The binding signals are exported into PLA to determine the relative binding response, elative affinity and relative binding for samples are also reported relative to the TG-1101 (TG Therapeutics, Inc.) reference standard. This test method is a validated assay, CTSOP477, used for TG-1101 (TG Therapeutics, Inc.) drug substance release testing. (b) Results
[00457] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Table 15, among the anti-CD20 antibodies tested, TG-1101 (TG Therapeutics, Inc.) has the highest binding affinities to both FcyRIIIa 158V and FcyRIIIa 158F receptors. Gazyva ranks the 2nd in binding affinities. For the high affinity receptor FcyRIIIal58V, TG-1101 (TG Therapeutics, Inc.) has ~15 folder higher affinity than Ocrevus; for the low affinity receptor FcyRIIIal58F TG-1101 (TG Therapeutics, Inc.) has ~10 folder higher affinity than Ocrevus. Shown in Table 16 are relative affinity and relative binding values using TG-1101 (TG Therapeutics, Inc.) ref. std. as reference. Results show that TG-1101 (TG Therapeutics, Inc.) has higher relative binding and relative affinity than all the other anti-CD20s.
Table 15: KD for FcyRIIIa 158V and FcyRIIIa 158V Binding by SPR
Figure imgf000130_0001
Table 16: Summary of Relative Affinity and Relative Binding Results
Figure imgf000130_0002
8.10 Example 10 — Fc Receptor Binding by Octet
(a) Materials and Method
[00458] This analysis was performed by LakePharma. Binding characterization were carried out on Octet HTX instrument at 25°C. Human Fc receptor panel were loaded onto Anti-Penta His (HIS IK) biosensors. Loaded sensors were dipped into a serial dilutions of the testing samples (300 nM start, 1 :3 dilution, 7 points). Kinetic constants were calculated using a monovalent (1 :1) binding model.
(b) Results
[00459] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Table 17, among the anti-CD20 antibodies tested, TG-1101 (TG Therapeutics, Inc.) has the highest binding affinities to both FcyRIIIa 158V and FcyRIIIa 158F receptors, corroborating the SPR data. Binding affinities to FCRN, which can affect PK, are similar among all the anti-CD20s. Compare to Ocrevus, TG-1101 (TG Therapeutics, Inc.) also has higher affinities for FcyRIIA and FcyRIIIB.
Table 17: Summary of Ko results against Fc Receptors
Figure imgf000131_0001
8.11 Example 11 — Clq Binding Assay
(a) Materials and Method
[00460] The Clq binding assay used for this analysis is an ELISA assay. Samples were coated on ELISA plates, HRP conjugated human Clq is incubated with samples on the plate. The bound HRP, in the presence of substrate TMB, generates a colorimetric signal. A 7-point dilution series of Test Materials were prepared in the concentration range of 15.00 ug/ml - 0.12 ug/ml with a dilution factor of 2. Sample dilutions are coated onto ELISA plates, and the plates incubated for 1 hour ± 30 min at room temperature. After coating, the plates are washed, blocked, and washed again. Peroxidase conjugated Clq was added, and the plates incubated for 1.5 hours ± 30 minutes at room temperature. After incubation and washing, a tetramethylbenzidine (TMB) substrate solution was added and plates incubated at room temperature for 7 minutes (-1 min/±30 seconds). This produces a colorimetric reaction which is proportional to the level of Clq bound. The reaction is stopped with the addition of IM sulfuric acid and the color is measured at 450 nm using a Molecular Devices SpectraMax microplate reader. SoftMax Pro is used to analyze the data with weighted nonlinear regression using a 4-parameter logistic fit. Binding activity results are reported as percentage potency relative to the TG-1101 (TG Therapeutics, Inc.) reference standard. EC50 of each test sample, based on the 4-parameter logistic fitted curve, were generated for additional information. This test method is a validated assay, CTSOP455, used for TG-1101 (TG Therapeutics, Inc.) drug substance and drug product release and stability testing.
(b) Results
[00461] Samples tested include TG-1101 (TG Therapeutics, Inc.), Gazyva, Rituxan, Ocrevus, and Arzerra. TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808) was used as control, As shown in Figure 10, Gazyva has minimal Clq binding (as expected), and the other 4 anti-CD20s exhibit dose dependent Clq binding. TG-1101 (TG Therapeutics, Inc.) has slightly higher maximum binding. The Clq binding activity expressed as percentage of the TG-1101 (TG Therapeutics, Inc.) reference standard is shown in Table 18. The ED50s of each anti-CD20 are also shown in Table 18, EC50 of TG-1101 (TG Therapeutics, Inc.) is slightly lower than the all anti-CD20s, suggesting TG-1101 (TG Therapeutics, Inc.) has higher affinity for Clq.
Table 18: Clq Binding Activity and EC50
Figure imgf000133_0001
8.12 Example 12 — Human Whole Blood B-Cell Depletion Assay
(a) Material and Method
[00462] TG-1101 (TG Therapeutics, Inc.) was characterized in an autologous normal human whole blood B-cell depletion assay in comparison to Gazyva, Rituxan, Ocrevus, and Arzerra. Whole blood from three human donors was used, and donors were selected to have the 158V/158V genotype for FCGR3A_SNP target (rs396991). Sample dilutions, in the concentration range of 0.000001 - 100 pg/ml, was added to the whole blood and incubated at 37°C for 24 hours in a humidified cell incubator. Aliquots of the blood were stained for markers including CD45 (lymphocyte population), CD3 (T cells), CD19 (B cells), and CD20 (B cells). B- cell depletion was evaluated by displaying cells in the CD45-positive lymphocyte gate, and enumerating CD3-positive T cells, CD19-positive B-cells, and CD20-positive B-cells. Percent of B-cell depletion (100-([100/B-/T-cell ratio in sample without antibody] x [B-/T-cell ratio in sample containing antibody])) was calculated and plotted against sample concentration.
(b) Results
[00463] Samples tested include TG-1101 (TG Therapeutics, Inc.) commercial reference standard RS-117808 (117808), A drug substance batch manufactured by the commercial process (C2) at Samsung Biologies (PPQ1), Gazyva, Rituxan, Ocrevus, and Arzerra. As shown in Figures 11A-C, all samples exhibited dose-dependent B-cell depletion activity, even though there are some minor donor to donor differences. Overall, TG-1101 (TG Therapeutics, Inc.) and Gazyva have higher B cell depletion activities than Arzerra, Rituxan, and Ocrevus. Three anti- CD20 antibodies and one anti-CD19 antibody were used in this experiment. The B-cell depletion ED50s calculated using each of the antibodies for B cell labeling are summarized in Table 19. Overall, TG-1101 (TG Therapeutics, Inc.) and Gazyva also have lower EC50 than Arzerra, Rituxan, and Ocrevus. On average, EC50 of TG-1101 (TG Therapeutics, Inc.) is more than tenfold lower than Ocrevus.
Table 19: Summary of ED50 of B Cell Depletion from Multiple B Cell Markers
Figure imgf000134_0002
8.13 Example 13 — Calculation of Pharmacokinetic (PK) Values
[00464] Relevant steady state PK parameters were calculated according to methods known in the art. A steady-state is reached when the quantity of drug eliminated in the unit of time equals the quantity of the drug that reaches the systemic circulation in the unit of time. Consequently, the half-life represents the time required to reduce the plasma concentration of the drug reached in steady-state by 50%. Wherein; t=Time, Vd= volume of distribution, and Cl= clearance. The half-life was calculated with the following formula:
Figure imgf000134_0001
[00465] The AUC is representative of the total dosage of the drug exposure over time. AUC is utilized as a metric when determining the formulations of an equivalent dosage, and their resulting tissue or plasma exposure. AUC is equivalent to the average concentration over a time interval. Wherein t=Time, and Cpt= the last measured drug concentration with respect to time. The AUC was calculated with the following formula:
Figure imgf000135_0001
[00466] The Cmax was obtained by the measurement of the highest point during the time of observation of drug concentration following the end of the absorption phase and the beginning of the elimination phase. The Cmin was obtained by the measurement of the highest point during the time of observation of drug concentration following the end of the absorption phase and the beginning of the elimination phase.
8.14 Example 14 — Methods for Determining Population Pharmacokinetic (PPK) Values for TG-1101 For Treatment of Autoimmune Disorders
[00467] TG-1101 serum concentration-time, dose, demographic, and covariate data from one Phase 2 study (TG1101-RMS201) and two Phase 3 studies (TG1101-RMS301 and TGI 101- RMS302) in subjects with RMS were pooled for the Pop PK analysis of TG-1101. The dataset was combined with a previous dataset of TG-1101 in subjects with hematologic malignancies.
[00468] All subjects in Studies TGI 101-RMS201, TGI 101-RMS301 and TGI 101-RMS302 with at least one TG-1101 dose administration were included in the dataset for the PK analysis. Subjects that did not have at least one quantifiable post-dose TG-1101 concentration were included in the dataset and flagged. Exposures for these subjects were determined based on typical population PK parameters.
[00469] In the Phase 2a clinical trial, TGI 101-RMS201, TG-1101 was administered as a single agent and compared with placebo to examine the level of B cell depletion by TG-1101 as well as determine the optimal dose and infusion time for TG-1101 in subjects with RMS. Based on the results of this study, a dose of 150 mg (infused over 4 hours) on Week 1 Day 1 followed by a dose of 450 mg (infused over one hour) on Week 3 Day 15 resulted in a median of >99% B cell depletion that was achieved on Week 4 and sustained until Week 24. The dosing regimen was well tolerated by the subjects with infusion-related reactions (Grade 1 and 2) being the commonly reported adverse event. Two Phase 3 studies in subjects with RMS have been completed. Two phase 3 studies, TGI 101-RMS301 (aka ULTIMATE I) and TGI 101-RMS302 (ULTIMATE II), were randomized, double-blind, double-dummy, active-controlled studies of TG-1101 compared with oral teriflunomide to assess ARR, safety and tolerability in subjects with RMS.
[00470] Overall, the dataset for PopPK analysis included a total of 8672 PK samples collected from 931 subjects. Pre-dose samples accounted for 10.02% of PK samples and records with missing information or outliers (> 10 standard deviations from the mean TG-1101 concentration at the nominal dosing time) accounted for 0.20% and were excluded. Post-dose samples that were BLQ accounted for 3.47% of the data. Exclusions resulted in no quantifiable post-dose PK samples for 36 subjects. Consequently, the PK analysis dataset included 7485 PK samples from 895 subjects, of which 5624 PK samples were from 591 subjects with RMS.
[00471] The final PopPK parameter estimates are presented in Table 20. PK parameter estimates for a typical subject (defined as a male subject that is ADA negative with a body weight of 73 kg from North America or Western Europe) were as follows: CL was estimated to be 11.6 mL/h, with IIV of 38.1%; Ve was estimated to be 3.18 L(IIV=15.0%); Vp was estimated to be 3.60 L (IIV=21.3%); and Q was estimated to be 11.6 mL/h.
[00472] Body weight and ADA were found to be statistically significant predictors of TG- 1101 CL. TG-1101 CL was modestly increased by 14% in subjects that were ADA positive compared to those had no quantifiable ADA. Lor the wide range of body weight in the RMS subpopulation (45.1 to 154 kg), CL ranged from 22% lower to 48% higher compared to that for a typical subject with a body weight of 73 kg. In addition at late times (417 d after the start of treatment), CL was reduced by a median of 12.5%.
[00473] Body weight, sex and region were found to be a statistically significant predictor of Vc- Subjects from Eastern Europe were found to have slightly higher (10%) Vc than Western Europeans and North Americans and females had slightly lower (7%) Vc than males. Lor the wide range of body weight in the RMS subpopulation (45.1 to 154 kg), V c ranged from 19% lower to 38% higher than that for a typical subject with a body weight of 73 kg.
[00474] After inclusion of body weight in the model, there was no effect of age, hemoglobin concentration, platelet count, white blood cell count, renal impairment or hepatic impairment on TG-1101 PK. Table 20: Parameter Estimates of the Final TG-1101 PK Model
Figure imgf000137_0001
Abbreviations: CL - clearance; CV% - percent coefficient of variation; ETA - individualspecific random effect; IIV - inter-individual variability; Q - inter-compartmental clearance; RSE - relative standard error; SD - standard deviation; SE - standard error; U2 - TG-1101 + umbralisib; Vc - volume of the central compartment; Vp - volume of the peripheral compartment. a The RSE of the parameter estimate is calculated as 100 x (SE/typical value); the RSE of the IIV magnitude is calculated as 100 x (SE/variance estimate). b Estimates for random effects and IIV are presented in CV% and based on the estimated values, calculated as ^variance x 100. c Shrinkage (%) as calculated as 100 x (1 -SD[ETA]A/ [variance]). dThe correlation coefficient between CL and Vc was estimated as 0.434. [00475] In the evaluation of the final TG-1101 PopPK model, standard errors were obtained and presented together with the parameter estimates in Table 20. Model parameters were precisely estimated with RSE < 20% for structural and covariate model parameters and RSE < 30% for random effects estimates. Shrinkage was acceptable for CL and Vc (5.32% and 32.6%, respectively) with large shrinkage on Vp (40%).
[00476] Key Goodness-of-fit (GOF) diagnostics for the TG-1101 final PopPK model suggested satisfactory fit with minimal bias in residuals over time and across predicted concentration values and showed good agreement between predicted and observed concentrations. Relative to the base model, ETA - covariate relationships were resolved and no further trends between ETAs (on CL and Vc) and covariates were evident in the RMS subpopulation, suggesting that the model adequately captured significant covariate relationships. Overall, the prediction-corrected visual predictive check (pcVPC) plots suggests that the model well predicts the central tendency of the observed TG-1101 concentrations and adequately captures the range of the data.
[00477] The relative importance of covariate effects included in the final PoPK model was evaluated with a forest plot of relative changes in exposure (Cmax,ss, Cmin,ss, and AUCss) when covariates were varied one at a time (i.e., univariate analysis). The effects of these covariates, including body weight, sex, region ADA and the fractional change in CL at late times, on TG-1101 exposures fell within the range of O. 8 to 1. 25 compared to the reference exposure (defined as the exposure for a male subject from North America/Western Europe with a body weight of 73 kg, who is ADA negative and has been on treatment for < 416 days. Consequently, none of the covariates were deemed clinically relevant. Furthermore, the covariates did not have a significant impact on the magnitude of IIV on CL or Vc. The combined effects of body weight and ADA reduced IIV by only 2.8%, from 39.2% in the base model to 38.1 % (Table 20) in the final model. Similarly, body weight, sex and region reduced IIV by 18.5% in Vc from 18.4% in the base model to 15.0% in the final model.
[00478] Figure 12 shows the Goodness-of-Fit (GOF) diagnostics of the TG-1101 final model. Figure 13 shows the pcVPC for the TG-1101 final PK model by study. Figure 14 shows the forest plot of covariate effects on TG-1101 drug exposure.
[00479] The final PopPK model was re-estimated with the dataset including excluded outliers. All structural parameters were precisely estimated with no relevant change in those estimates, while the IIV terms were inflated with the inclusion of the outliers. It can therefore be expected that exclusion of outliers during the model development eliminated the influence of these outliers and minimized spurious findings in the covariate analysis. The final PopPK model was also re- estimated including the RMS subpopulation only. The fit of the model to the RMS data resulted in similar structural parameters and random effects, with the exception of marginally lower IIV on CL; precision of parameter estimation was also similar.
[00480] Conclusions: TG-1101 PPK Analysis
[00481] The final model of TG-1101 was utilized to obtain individual post hoc estimates of PK parameters. For each subject enrolled in studies TGI 101-RMS201, TGI 101-RMS301, TGI 101-RMS302, PK parameters (AUCss, Cavg,ss, Cmax,ss, and Cmin,ss) at Week 48 were estimated based on post hoc PK parameters.
[00482] The geometric mean /2 (90% confidence interval [CI]) was calculated to be 21.8 days (21.4, 22.1 days). Median time to reach steady state was determined to be 15. 5 weeks. Accordingly, there was no accumulation for subjects that received the per- protocol regimen of 150 mg TG-1101 on Day 1 followed by 450 mg on Day 15, Week 24 and Week 48. The median Cmax ratio at Week 24 to Cmax on Day 1 was 3.04 (range 3.00 to 3.42) consistent with the 3- fold increase in the amount of the dose and indicative of no accumulation. Similarly, the Cmax ratio at Week 48 to Week 24 was 1, indicative of no accumulation.
[00483] The model-predicted geometric mean AUCss, Cavgss, Cmaxss, and Cminss was 3000 ug d/mL (±28%), 8940 ng/mL (±28%), 139000 (±15.1%), and 139 (±170%), respectively.
8.15 Example 15 — Commercial-Scale Manufacturing Process of Making TG-1101 in YB2/0 Rat Hybridoma Cells
[00484] In this Example, the manufacturing process for TG-1101 expressed in YB2/0 rat hybridoma cells at 15,000 L is described. An overview of the manufacturing process for TG- 1101 is illustrated in the flow diagram of Table 21.
Table 21: TG-1101 Manufacturing Process Flow Diagram
Figure imgf000140_0001
[00485] In summary, the production of each batch of TG-1101 began with the thaw of a working cell back (WCB) vial, described further below. The culture was expanded through a series of shake flasks and seed bioreactors to meet the inoculum requirements of the 15,000 L production bioreactor, which was operated in fed-batch mode. The bioreactor was harvested and clarified by centrifugation followed by depth filtration. The clarified harvest was purified by three chromatography steps including Protein A, cation exchange, and anion exchange, designed to purify the TG-1101 and to reduce process impurities such as host cell protein and residual DNA. The purification process (as illustrated in Example 4) contained steps to ensure viral safety including viral inactivation (solvent/detergent) and a viral filtration step. Final UFDF and formulation steps are used to concentrate and buffer exchange TG-1101 into the formulation buffer and to the desired product concentration. The ready to fill drug substance was formulated to obtain TG-1 lOlat a concentration of 25.0 mg/mL in 25 mM sodium citrate, 154 mM sodium chloride, 0.07% polysorbate 80, pH 6.5. After filling, the TG-1101 drug substance was frozen at <-60°C and then stored frozen at <-35 °C.
[00486] Additional descriptions of the upstream and downstream process operations are provided below.
(a) Expresion Vector, Production Cell Line, and Cell Banks
[00487] The host cell line used for generation of the TG-1101 producing cell line was the rat cell line YB2/0. The production cell line, R603-12D11, was developed after transfecting the expression vector HK463-25 (containing the immunoglobulin heavy and light chain cDNA sequences of TG-1101) into the YB2/0 host cell line. Figure 15 depicts the expression vector map of HK463-25 to produce TG-1101, in a 15,000 L bioreactor.
Expression Vector
[00488] The expression vector HK463-25 included various elements that were optimized for stable expression in the YB2/0 host cell line. The Rous Sarcoma Virus Long Terminal Repeat (RS V LTR) promoter was used for the constitutive expression of both heavy and light chain cDNAs. This promoter corresponds to the Long Terminal Repeat of the RSV genome which contains enhancer elements in its 5’ region and has strong transcriptional activity in the YB2/0 cell line. Transcriptional termination and polyadenylation of both heavy and light chain cDNAs were provided by the human growth hormone polyadenylation sequence (hGH poly A). A chimeric intron was introduced 5’ to the cDNA sequence of each antibody chain to improve expression. This intron was optimized for splicing and is composed of a 5’ donor sequence from human beta-globin and a 3 ’ acceptor sequence from an Ig heavy chain variable gene. The beta lactamase gene conferred ampicillin resistance (AmpR) and was provided to enable production of the plasmid in E. coli. The enzyme neomycin-phosphotransferase II (NeoR) was under the control of the SV40 promoter and confers resistance to the transfected cell line to the antibiotic G418, thus acting as a selectable marker. Dihydrofolate reductase (Dhfr) was under the control of the SV40 promoter and conferred resistance to methotrexate (MTX) and can also act as a selectable and amplifiable marker in the transfected cell line.
[00489] The HK463-25 expression vector (Figure 15) was 11.1 kb in size and contained five open reading frames for the antibody heavy chain, light chain, Dhfr, NeoR and AmpR genes in the same orientation. The restriction sites shown in the figure were used for Southern blot analysis of the integration of the construct. A unique Notl restriction site located 3’ to the NeoR gene was used for linearization of the vector prior to transfection.
The production cell line, R603-12D11
[00490] The production cell line, R603-12D11, was generated after transfection of the host cell line, selection and screening of transfectants, and then limiting dilution cloning. The clones were screened, production cell line R603-12D11 was selected, and adapted to serum free medium. A pre-seed stock (PSS) cell bank was prepared. An overview of the steps involved in the generation of production cell line R603-12D11 is shown in Table 22.
Table 22: Production Cell Line R603-12D11 Generation
Figure imgf000143_0001
[00491] A cryovial of the YB2/0 cell bank (YB2/0-301 04/147) was thawed and the cells were grown by dilution to a cell density of 1 * 105 cells/mL every 3 to 4 days with fresh culture medium (EMS medium with 5% FCS). The cells were seeded at a density of 2 105 cells/mL on the day before transfection in order to reach exponential phase prior to transfection. 44.5 pg of the NotLlinearized expression vector HK463-25 (Figure 15) was transfected into 5/ 106 cells by electroporation using the animal component free Optimix reagent (Equibio). The cells were diluted in culture medium and seeded at 100 cells/well in 96-well plates. Selection with 1 g/L G418 in the culture medium was initiated three days after electroporation.
[00492] The transformants were initially screened for titer by ELISA after selection in the G418 medium. Over 3000 transformants were screened and over 200 of the best producing wells were selected for further testing and for continued passaging. A second titer screen was performed to further reduce the number of clones. This screen was followed by additional screening for antibody fucose level by ELISA. Fucose levels less than 40% were considered desirable in order to provide the expected level of activation of CD 16 by the antibody; CD 16 activation is inversely related to the fucose levels. Further screening using a cell-based CD 16 activation test with assessment of IL-2 secretion in addition to assessment of the free Kappa/IgG ratio (<0.2 for purification ease) resulted in narrowing to a total of 5 cell lines for further development. [00493] The 5 candidate cell lines were cloned by limiting dilution at 0.4 cells/well in EMS medium with 5% FCS. Clones were screened for IgG productivity, fucose levels, CD 16 activation, and free Kappa/IgG ratio to identify production cell line R603-12D11. As the selected cell line was expanded from the cloning step, it also underwent a transition into serum free media and was re-screened to ensure the desired phenotype before the preparation of a small cell bank. This small cell bank was then thawed and expanded to generate the R603-12D11 pre-seed stock cell bank (PSS). The R603-12D11 PSS cell bank was demonstrated to be free from mycoplasmal, adventitious virus and microbial contamination prior to the generation of the MCB.
[00494] Characterization of the master and working cell banks derived from the R603-12D11 production cell line, included testing for identity, genotypic and phenotypic features, as well as for the presence of adventitious agents.
Master Cell Bank (MCP) Preparation and Testing
[00495] The MCB was manufactured at Henogen (later acquired by NovaSep). The MCB lot G071/MCB/070208 was prepared by thawing and expanding one vial of the production cell line R603-12D11 pre-seed stock in serum-free medium EM-SF2 P500 H4 (EMS basal medium supplemented with 2-mercaptoethanol, ethanolamine, NaHCO, ferric citrate, pluronic acid, HEPES and recombinant human insulin). The cells were expanded in T-flasks and roller bottles for eleven days. The cell suspension was concentrated by centrifugation and aliquoted into 13 separate fractions. Each fraction was centrifuged and resuspended to a target cell density of 10* 106 cell/mL in freezing medium (90% EM-SF2 P500 H4 + 10% DMSO). The suspended fractions were then each aliquoted into 18 cryovials per fraction resulting in a total of 234 cryovials of the MCB. The cryovials were placed on dry ice and then into cryoboxes which were placed in a -80°C freezer for 21 hr. The cryovials were transferred into liquid nitrogen tanks on 19 February 2007 for long-term storage and are currently stored in multiple locations. The number of cell generations from the PSS of the production cell line to the MCB is 10.4.
[00496] Following the generation of MCB lot G071/MCB/070208, direct testing of the MCB, as well as further characterization by testing a WCB derived from it, were performed. Identity testing of the MCB was performed; testing results confirm the identity of the MCB as rat derived. Performance qualification was confirmed by thawing a vial of the MCB and monitoring for cell viability. Assessments of copy number, restriction endonuclease profile, number of integration sites, integrity of RNA coding sequence, and RNA quantitation were conducted. The number of heavy and light chain copies integrated into the genome was estimated at 1.13 and 2.14, respectively, by quantitative polymerase chain reaction (Q-PCR). Southern blot assessment of the plasmid integration site demonstrated comparable hybridization patterns for the MCB and PSS. The number of integration sites for both the MCB and WCB was similarly measured to be 1 in both cell banks via Southern blot. In addition, intra-chromosomic integration of the HK463-25 expression plasmid with insertion at a single locus of a metacentric chromosome was demonstrated by fluorescence in situ hybridization (FISH) analysis. For the MCB, integrity of the RNA coding sequence was consistent with the reference sequence, and RNA quantitation was consistent with the PSS cell bank for both heavy and light chains.
[00497] Next generation nucleic acid sequencing (NGS) using targeted locus amplification (TLA) was performed (Cergentis, Utrecht, Netherlands) on the MCB to confirm that the expressed TG-1101 antibody has the correct amino acid sequence, and to also interrogate for low levels of sequence variants. Adventitious viral, mycoplasmal and microbial agent testing of the MCB was carried out and acceptance criteria established for release of the MCB including those for adventitious viral, mycoplasmal and microbial agents were met. The collective MCB testing results confirmed the suitability for the establishment of the MCB.
Working Cell Bank Lot G140/R603/WCB001
[00498] The first WCB was manufactured at NovaS ep (after acquiring Henogen). To prepare the WCB, one vial of MCB G071/MCB/070208 was thawed and expanded in serum-free medium EM-SF2 P500 H4 in flasks and roller bottles over eleven days. The expanded cell suspension was concentrated by centrifugation and aliquoted into 22 identical fractions. Each fraction was centrifuged and resuspended to a target cell density of 12.1 106 cell/mL in freezing medium (90% EM-SF2 P500 H4 + 10% DMSO). The suspended fractions were then each aliquoted into 18 cryovials resulting in a total of 396 cryovials of the WCB. The lot was designated as G140/R603/WCB001. The cryovials were placed on dry ice and then into cryoboxes which were placed in a -80°C freezer for 24 hr. The cryovials were transferred into liquid nitrogen tanks for long-term storage on 22 September 2009. The WCB is stored in at least two different storage sites including smaller numbers of vials stored for shorter duration at the manufacturer. The number of cell generations from the PSS of the production cell line to the WCB is 21.4.
[00499] Testing of WCB lot G140/R603/WCB001 was performed including identity testing. Testing results confirmed the identity of the WCB as rat derived. Performance qualification was confirmed by thawing a vial of the WCB and monitoring for cell viability, doubling time, and IgG productivity. Assessments of copy number, restriction endonuclease profile, and number of integration sites, were conducted. The number of heavy and light chain copies integrated into the genome was estimated at 1.2 and 2.5, respectively, by quantitative polymerase chain reaction (Q-PCR). These results are consistent with that of the MCB. Southern blot assessment of the plasmid integration site demonstrated comparable hybridization patterns for the WCB and MCB. The number of integration sites for the WCB was measured to be 1 via Southern blot.
[00500] Next generation nucleic acid sequencing (NGS) using targeted locus amplification (TLA) was performed (Cergentis, Utrecht, Netherlands) on the WCB to confirm that the expressed TG-1101 antibody has the correct amino acid sequence, and to also interrogate for low levels of sequence variants. Adventitious viral, mycoplasmal, and microbial agent testing was conducted and all acceptance criteria established for release of the WCB including those for adventitious viral, mycoplasmal and microbial agents were met. The collective WCB testing results confirmed the suitability for the establishment of the WCB.
Phenotypic Characterization of Cell Banks
[00501] Cell line phenotypic stability studies were performed for the MCB and two WCBs generated for manufacturing of TG-1101. One vial each of the MCB, WCB lot G140/R603/WCB001, and a new WCB lot 127646-001 were thawed and passaged for approximately 60 generations. VCD, cell viability, and titer samples were taken at every passage. At intervals of approximately 15 cell generations, cells were cryopreserved as research cell banks (RCBs). After approximately 60 generations, vials from each RCB were thawed and expanded for 7 days. On day 7, shake flasks were inoculated, cultured under fed-batch conditions, and cultivated for 12 days. Daily sampling for VCD, cell viability, titer, specific productivity, and quality attributes of the harvested fed batch culture were used to assess the stability of each cell bank at its corresponding generation number. Comparability in cell growth, titer, and product quality was used as a general basis for determining phenotypic stability of the cell banks. In addition, specific productivity results that exceed 70% of the results from control cultures with low number of generations were used as a specific basis for determining phenotypic stability of the cell banks. The results for the three cell bank phenotypic stability studies are shown in Table 23 for MCB lot G071/MCB/070208, Table 24 for WCB lot G140/R603/WCB001, and Table 25 for WCB lot 127646.
Table 23: Phenotypic Stability Results of MCB Lot G071/MCB/070208
Figure imgf000147_0001
Table 24: Phenotypic Stability Results of WCB Lot G140/R603/WCB001
Figure imgf000148_0001
After WCB vial thaw to the start of production fedbatch culture
Table 25: Phenotypic Stability Results of WCB Lot 127646
Figure imgf000149_0001
[00502] Thawed MCB and WCB that were use during production of TG- 1101 at 15,000 L were tested for viable cell density and cell viability results to assess and confirm cell bank storage stability.
(b) Upstream Process and Process Controls
[00503] A process flow diagram of the upstream unit operations including operational controls and in-process controls are provided in Table 26. Among other controls, bioburden and endotoxin were measured in the batched media of the seed and production bioreactor stages. Table 26: Upstream Operational and In-Process Controls
Operational Controls Unit In-Process Controls
Figure imgf000150_0001
Figure imgf000150_0002
Cell Culture Media and Feeds Preparation
[00504] Cell culture growth medium (CDM4Mab) and feeds (BalanCD CHO Feed4, glucose feed, and glutamine feed) were prepared with Water for Injection (WFI). Media and feeds were filtered (<0.2 pm) into sterile vessels and stored prior to use as needed. A cholesterol lipid concentrate was supplemented into the CDM4Mab growth medium during preparation to support cell growth from the inoculum expansion stages through the production bioreactor. In addition, the cholesterol lipid concentrate was added to the production bioreactor as fixed bolus feed additions on process days 0 and 4. Operational and in-process controls for cell culture media and feeds are described in Table 27.
Table 27: Cell Culture Media and Feeds Preparation and Storage Controls
Figure imgf000151_0001
a Room temperature at SBL: 17-25°C b Operating temperature: 37.0°C (36.5-37.5°C)
Inoculum Expansion
[00505] Inoculum expansion steps included thaw of the WCB vial and growth in shake flasks and/or cellbags of increasing size and volume to provide sufficient cell mass to inoculate the seed bioreactor stages. The steps are performed by growth in inoculum expansion growth medium (CDM4Mab). To initiate the process, a vial of WCB G140/R603/WCB001 was thawed in pre- warmed water in a 37.0°C water bath. The thawed vial contents were transferred into prewarmed medium and diluted to achieve a target seed density of 0.55 x 106 viable cells/mL.
[00506] The culture was placed in an initial 125 mL shake flask and grown in a shaking incubator at 37.0°C/5.0% CO2 for 1 day. Every 2 to 3 days, the culture was expanded into shake flasks of larger volume and/or to multiple shake flasks. At each stage, a seeding density of 0.30 x 106 viable cells/mL was targeted. The final inoculum preparation stage consisted of a 50 L cellbag. After 2 to 3 days of growth, the viable cell density was checked and culture was further processed to the seed bioreactor stages. Operational and in-process controls for inoculum expansion are described in Table 28.
Table 28: Inoculum Expansion Controls
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
a Shaker platform throw radius dependent parameter. Shaker platform throw radius: 22 mm
Seed Bioreactors
[00507] The seed bioreactor stages further increase the volume and cell culture biomass prior to inoculation of the production bioreactor. The medium used in these stages was inoculum expansion growth medium (CDM4Mab). The seed bioreactor stages were 120 L, 600 L, and 3000 L stainless steel bioreactors. The bioreactors were equilibrated after media addition. Dissolved oxygen and pH probes were calibrated prior to use. Initial bioreactor set points included temperature, pH, dissolved oxygen, and agitation rate. Each bioreactor was inoculated with cell culture from the preceding stage and the culture was grown for 2 to 3 days. Seed bioreactor operational and in-process controls are summarized in Table 29.
Table 29: Seed Bioreactor Controls
Figure imgf000154_0002
Figure imgf000155_0001
Figure imgf000156_0001
Production Bioreactor
[00508] The production bioreactor stage is the final cell culture process stage which further increases the volume and mass of the cell culture for expression of the TG-1101 antibody with acceptable product quality. The basal medium used in this stage was growth medium (CDM4Mab) and feed additions were performed at specified days or criteria during the process. The production bioreactor was a 15,000 L stainless steel bioreactor.
[00509] The production bioreactor was equilibrated after media addition. Inoculum expansion medium was added to an initial volume target. Dissolved oxygen and pH probes were calibrated prior to use. Initial bioreactor set points included temperature, pH, dissolved oxygen, and agitation rate. The bioreactor was inoculated with cell culture from the N-l seed bioreactor at a viable cell density target of 0.5 x 106 viable cells/mL. During the production bioreactor process, pH was controlled with base addition and CO2 sparging on demand. Dissolved oxygen was controlled with oxygen and air sparging on demand. Antifoam was added as needed to mitigate foaming concerns. Offline pH, pCO2, pO2, osmolality, and metabolites were monitored daily as were viable cell density (VCD) and viability. The BalanCD CHO Feed4 was added on Days 3, 5, 7, 9 at specified volumes (4.0% of initial bioreactor working volume). A calculated volume of glucose solution was fed (to 4.00 g/L) when daily bioreactor sample measured values were <3.00 g/L on specified process days. A fixed volume of glutamine solution was bolus fed on Day 3 (3.0% of initial bioreactor working volume), and a calculated volume was fed (to 4.00 mM) when daily bioreactor sample measured values were <3.00 mM on specified process days. The production bioreactor was harvested based on either culture duration or cell viability criteria, whichever occured first. [00510] Unprocessed bulk samples were removed prior to the clarification unit operation for adventitious agent testing as described in Example 4. Production bioreactor operational and in- process controls are described below in Table 30.
Table 30: Production Bioreactor Controls
Figure imgf000157_0001
Figure imgf000158_0001
a Temporary excursions are allowed. b Agitation is dependent on volumetric scale and bioreactor impeller. Target (NOR) are equivalent to power/volume 50 (42-59) W/m3 at SBL. AR is equivalent to power/volume 42-70 W/m3. c Fed on Days 3, 5, 7, 9. d Fed on Days 4-7 as needed. e Glucose/glutamine are not measured after glucose/glutamine feed addition; values refer to target concentrations used in the calculation for determining the glucose/glutamine feed amount required. f Fed on Days 3-12 as needed. g Antifoam amount is calculated based on initial working volume of production bioreactor. Target is addition amount at one time. h Harvest occurs when either culture duration reaches its target/NOR, or when final cell viability drops lower than its NOR, whichever occurs first. As a result, it is possible for harvest to occur when final cell viability is below its NOR, or culture duration is below its NOR.
(c) Downstream Process and Process Controls
[00511] A process flow diagram of the downstream steps including operational controls and in-process controls are provided in Table 31. As shown in Table 31, in-process controls were incorporated into the process. Among other controls, bioburden and endotoxin were measured during multiple stages of the downstream process. TABLE 31: Downstream Operational and In-Process Controls
Operational Controls, Process Step In-Process Controls Target
Figure imgf000159_0001
Figure imgf000160_0001
a Beginning, middle and end of filling operation
Harvest Clarification
[00512] The cell culture supernatant was harvested and clarified from the 15,000 L bioreactor to remove cells and cell debris. The clarification was performed using a continuous centrifugation followed by depth filtration.
[00513] The harvest clarification step was operated in a room in which the controlled temperature range was 17-25°C; the harvest pool vessel was a jacketed tank, which maintains the pool at 2-8°C. The centrifuge shot interval was set based on the packed cell volume percentage (PCV) and adjusted to allow for 80% bowl fill. The flow rate to the centrifuge was actively controlled; the filtration feed flow was the same as the centrifuge feed flow. Process parameters included centrate back pressure, the depth filtration operating pressure, inlet pressure, harvest weight, bioburden and endotoxin.
[00514] The centrate was clarified by use of a three-stage filtration process as required to pass through a 0.2 pm filter (Millistak A1HC POD depth filters, 1.2/0.5 pm filters and 0.45/0.22 pm sterilizing grade filters). Before use, the filters were flushed with WFI followed by equilibration. The centrate was pumped through the filters, which are monitored to ensure acceptable back pressure. Air was used to expel the contents in the filters followed by a buffer flush. The clarified harvest was stored at 2 to 8°C for <11 days.
Protein A Column Chromatography (ProA)
[00515] Protein A column chromatography was performed using MabSuRe Select resin (Cytiva) in bind/elute mode. This step provided capture and purification of TG-1101 with reduction of process impurities such as cell culture components, HCP, and residual DNA, as well as for the provision of viral safety. [00516] The packed column was assessed for HETP performance using sodium acetate/benzyl alcohol buffer. During manufacturing, all column operations were performed at 13 to 25°C. Before loading, the column was sanitized with 0.5 M sodium hydroxide and flushed with WFI. The column was equilibrated with Equilibration Buffer (25 mM Tris, 25 mM NaCl, 5 mM EDTA, pH 7.1). The clarified harvest was mixed briefly and then loaded onto the column using a maximum of 36 g TG-1101/L resin load and the column was washed with Wash 1 Buffer (equilibration buffer) followed by a second wash with a high salt, Wash 2 Buffer (25 mM Tris, 1.2 M NaCl, 5 mM EDTA, pH 7.1), followed by an additional wash using Wash 3 Buffer (equilibration buffer). Bound TG-1101 was eluted 200 - 220 cm/h with elution buffer (25 mM sodium citrate, pH 3.6) using elution peak collection by A280, not to exceed 2.4 column volumes. The eluate was collected in a tank containing neutralization buffer (2.0M Tris, pH 7.5) and filtered via a 0.2 pm filter before transfer to a different tank. The Protein A column was sanitized with 0.5M sodium hydroxide. Up to three cycles per batch may be run; if multiple cycles were required for the Protein A process, the column was re-equilibrated for the next cycle with equilibration buffer. After sanitization, the Protein A column was neutralized with equilibration buffer and stored at 13 to 25°C in 200 mM sodium acetate, 2% benzyl alcohol, pH 5.0. The pooled (if more than one cycle), neutralized eluate was diluted with 5mM Sodium Phosphate, pH 7.2 to a concentration of < 10 g/L and stored at 13-25°C for <24 hours or at 2 to 8°C for <11 days.
Solvent Detergent Viral Inactivation (SDVI)
[00517] The Protein A capture chromatography step was followed by a solvent detergent viral inactivation (SDVI) step to inactivate potential viral agents. In the currently validated manufacturing process, the Protein A elution pool was diluted and treated with 3.5% (v/v) TnBP, 12% (w/v) polysorbate 80 and held at 24.0-26.0°C for at least 120 minutes while mixing. The SDVI pool was filtered with a 0.2 pm filter before transfer to a different tank, where it was diluted with 5 mM sodium phosphate, pH 7.2 to 50 mOsm/kg, and the pH adjusted to 7.2 as needed. After pH adjustment, the pool was held at 13 to 25°C for <30 hours before proceeding to the CEX column. Cation Exchange Chromatography (CEX)
[00518] Cation exchange column chromatography was performed using SP Sepharose Fast Flow (Cytiva) in bind/elute mode. This step provided further purification of TG-1101, removing residual process impurities (HCPs, DNA, residual polysorbate 80, and TnBP).
[00519] The packed column was assessed for HETP performance using a sodium acetate/benzyl alcohol-containing buffer. During manufacturing, all column operations were performed at 13 to 25°C. In the currently validated manufacturing process, before loading, the column was sanitized with 0.5 M sodium hydroxide and rinsed with WFI. The column was equilibrated using equilibration buffer (20 mM sodium phosphate, pH 7.2). The viral inactivated/diluted solution was loaded onto the column at a maximum of 65 g/L resin load. The column was washed with Wash 1 Buffer (equilibration buffer) followed by a second wash with Wash 2 Buffer (equilibration buffer in the reverse direction). Bound TG-1101 was eluted using 20 mM sodium phosphate, 150 mM NaCl, pH 7.2 and elution peak collection by A280 monitoring. The eluate was filtered (0.2 pm) and stored at 13 to 25°C for < 72hours or at 2 to 8°C for <11 days. After elution, the column was stripped with 2 M NaCl followed by sanitization with 0.5 M sodium hydroxide. One cycle per batch was allowed. After completion, the column was sanitized (0.5 M sodium hydroxide) and stored in storage buffer (200 mM sodium acetate, 2% benzyl alcohol, pH 5.0).
Anion Exchange Membrane Chromatography (AEX)
[00520] Anion exchange membrane chromatography was performed using a Mustang Q (Pall Corporation) membrane absorber (MA) filter in flow-through mode. This step provided further purification of the TG-1101; the product flowed through the membrane and remaining impurities (DNA, HCP and viruses) were retained on the membrane. The membrane was single use (i.e., each individual membrane cannot be reused) and several membrane capsules may be used per batch at the appropriate loading level.
[00521] To perform the step, in the currently validated manufacturing process, eluate from the cation exchange column chromatography step was diluted with 20 mM sodium phosphate, pH 8.0, followed by the adjustment of the pH to 8.0. The concentration was determined and the number of cycles calculated based on protein concentration such that the loading was 200 to 700 g TG-1101/L membrane load. The membrane was sanitized with 0.5 M sodium hydroxide, flushed with 2 M NaCl then with WFI, before being equilibrated with Equilibration Buffer (20 mM sodium phosphate, 75 mM NaCl, pH 8.0) in preparation for the load. After loading, the membrane was chased with 20 mM sodium phosphate, 75 mM NaCl, pH 8.0, to maximize recovery. The collected flow- through containing product from all cycles was filtered (0.5/0.2 pm), diluted to <6 g/L with 75 mM sodium citrate, 312 mM NaCl, pH 6.0, and the pH is adjusted to 6.8. The adjusted AEX pool was stored at 13 to 25°C for < 72 hours or at 2 to 8°C for <11 days.
Viral Filtration (VF)
[00522] Viral filtration was performed by filtering through a Viresolve prefilter in series with a Viresolve Pro viral filter (Millipore Sigma), at an operating temperature Target Range of 13- 25°C. The process used sufficient filters to meet the loading limit. The step was designed to remove potential viruses, including small viruses such as parvovirus.
[00523] To perform the filtration, the filters were set up in series and flushed with WFI, integrity tested, then sanitized with 0.5 M sodium hydroxide. This was followed by flushing with equilibration buffer (25 mM sodium citrate, 154 mM NaCl, pH 6.5). The filtered Mustang Q membrane flow-through was processed through the viral reduction filters.
[00524] Protein concentration was determined and used to confirm the membrane load ratio was <600 g/m2 After loading, the membrane was chased with equilibration buffer and post-use integrity testing was performed. Viral filtrate was stored at 13 to 25°C for <72 hours or at 2 to 8°C for <11 days.
Ultrafiltration / Diafiltration (UFDF) Controls
[00525] The UFDF step was used to concentrate the viral filtrate and buffer exchange into diafiltration buffer at an operating temperature Target Range of 13-25°C. The process used a tangential flow filter with a 30 kDa molecular weight cut-off. The setup included up to sufficient filters to meet the loading limit of < 250 g/m2. To perform the operation, the membranes were sanitized with 0.5M NaOH, flushed with WFI, and flushed/equilibrated with diafiltration buffer before the start of the unit operation.
[00526] The unit operation included an initial ultrafiltration (UF1) step in which the TG-1101 was first concentrated to a target of 39 mg/mL. This was followed by diafiltration (DF) into diafiltration buffer (25 mM sodium citrate 154 mM NaCl, pH 6.5) with 8 diavolumes with an upper limit of < 9.2 diavolumes. The ultrafiltration system was flushed with diafiltration buffer to maximize recovery; the flush was transferred to the formulation vessel and combined with the UF/DF Pool. The membranes were flushed with WFI, cleaned with 0.5M NaOH, 250 ppm sodium hypoclorite, flushed with WFI, and stored in 0.1 M sodium hydroxide. The diluted UFDF pool was stored at 15 to 25°C for <18 hours.
Formulation, Filtration, and Filling
[00527] The formulation step included the addition of concentrated polysorbate 80 in formulation buffer to achieve the final drug substance formulation, at an operating temperature Target Range of 17-25°C. The step was performed by adding the stabilization buffer (25 mM sodium citrate 154 mM NaCl, 10 g/L polysorbate 80, pH 6.5). After the addition of the stabilization buffer, the pool was diluted to a target of 23.5 to 26.5 mg/mL with 25 mM sodium citrate 154 mM NaCl, 700 mg/L polysorbate 80, pH 6.5, resulting in a ready-to-fill drug substance in the formulation buffer of 25 mM sodium citrate, 154 mM NaCl, 0.07% polysorbate 80, pH 6.5.
[00528] The formulated bulk drug substance was transferred and 0.2 pm filtered into 6 L Celsius® FFT bags in a closed, single use system to a target fill volume of 5.50 L. After filling, the formulated bulk drug substance was frozen at <-60 for >17 hours and stored at < -35°C.
8.16 Example 16 — Onset and Maintenance of No Evidence of Disease Activity (NEDA) With Ublituximab
[00529] This Example describes evaluation of the timing of onset of no evidence of disease activity (NEDA) and proportion of participants maintaining NEDA with ublituximab in pooled post hoc analyses of ULTIMATE I and II. ULTIMATE I (NCT03277261) and ULTIMATE II (NCT03277248) were identical, Phase 3, randomized, multicenter, double-blind, active control studies that evaluated the efficacy and safety of ublituximab vs teriflunomide in participants with relapsing multiple sclerosis (RMS). Ublituximab is a novel monoclonal antibody targeting a unique epitope of CD20. Ublituximab is glycoengineered for enhanced antibody-dependent cellular cytotoxicity and is administered in 1-hour infusions after the first infusion. In ULTIMATE I and II, ublituximab significantly improved annualized relapse rate as well as number of gadolinium-enhancing (Gd+) T1 lesions and new/enlarging T2 lesions, and a higher proportion of participants achieved 3 -parameter NEDA (NEDA-3) rates with ublituximab vs teriflunomide. In pooled post hoc analyses, NEDA rates were 44.6% vs 12.4% at Weeks 0-96 for ublituximab vs teriflunomide cohorts, respectively, and 82.1% vs 22.5% at Weeks 24-96 (rebaselined; PO.OOOl for both). During Weeks 24-96 (re-baselined), 17.9% of ublituximab- treated participants had evidence of disease activity (EDA), and relapse was the most common component (11.4% ublituximab vs 22.9% with teriflunomide). In contrast, 77.5% of teriflunomide-treated participants had EDA, and the most common component was new/enlarging T2 lesions (71.6% with teriflunomide vs 3.1% with ublituximab).
(a) Method
[00530] The Phase 3 ULTIMATE I (N=549) and II (N=545) studies evaluated ublituximab 450 mg intravenous infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) or teriflunomide 14 mg oral once daily for 96 weeks in participants with RMS. Clinical evaluations were performed every 12 weeks, and magnetic resonance imaging (MRI) assessments were performed at weeks 24, 48, and 96. Pooled post hoc analyses evaluated the proportion of participants achieving and maintaining NEDA at Weeks 0-24, 24-96, 24-48, and 48-96. NEDA (e.g., NEDA-3) was defined as no confirmed relapses, no Gd+ T1 lesions, no new/enlarging T2 lesions, and no 12- week confirmed disability progression. The results are described in Figures 16A-16C.
(b) Results
[00531] Among ublituximab-treated participants, 53.4% achieved NEDA during Weeks 0-24, and 45.2% of participants achieved NEDA during Weeks 0-24 and maintained NEDA during Weeks 24-96. In addition, 36.9% of participants had EDA in Weeks 0-24 but then achieved NEDA in Weeks 24-96, bringing the total proportion of participants with NEDA to 82.1% for Weeks 24-96 (re-baselined; Figure 16A). After approximately 6 months of treatment, 93.6% of participants achieved NEDA during Weeks 24-48, and 83.5% of participants achieved NEDA during Weeks 24-48 and maintained NEDA during Weeks 48-96. An additional 4.8% of participants had EDA during Weeks 24-48 but achieved NEDA in Weeks 48-96, for a total of 88.2% of participants with NEDA during Weeks 48-96 (Figure 16B). The leading cause of disease activity during Weeks 0-24 was new/enlarging T2 lesions (occurring in 42.4% of participants). However, this MRI activity decreased in Weeks 24-48 and continued to decrease in Weeks 48-96 (Figure 16C).
(c) Conclusions
[00532] Ublituximab treatment in the Phase 3 ULTIMATE studies resulted in a high proportion of participants achieving and maintaining NED A. More than half of participants (53.4%) achieved NEDA by Week 24, which increased to more than 82% at Week 96. The majority of participants maintained NEDA once achieved.
8.17 Example 17 — Characterization of Cytopenias with Ublituximab in the ULTMATE I and II Phase 3 Studies in Participants with Relapsing Multiple Sclerosis
[00533] This Example describes characterization of cytopenias with ublituximab in the ULTIMATE I and II studies. Along with sustained B-cell depletion, pharmacodynamic (PD) studies of ublituximab have reported a transient decline in the percentage of total T cells and NK cells as well as a reciprocal increase in myeloid cells following the initial dose of ublituximab at Day 2 (Fox EJ, et al. Presented at: ACTRIMS (February 24-26, 2022), West Palm Beach, FL, Poster P105; Lovett-Racke AE, et al. J Neuroimmunol. 2021, 359:577676). Lymphocyte counts at Day 2 have not been evaluated in pivotal trials of other anti-CD20 agents (Hauser SL, et al. N Engl J Med. 2017, 376(3):221-234; Hauser SL, et al. N Engl J Med. 2020, 383(6): 546-557). Additional evaluations of ULTIMATE I and II data were conducted to understand the incidence and kinetics of cytopenias with ublituximab as well as potential association with infections. In the Phase 3 ULTIMATE I and II studies in participants with RMS), adverse events with ublituximab overall were not imbalanced compared with teriflunomide and were consistent with the anti-CD20 class.
(a) Method
[00534] The ULTIMATE I and II enrolled a total of 1094 adults from 10 countries with a diagnosis of RMS (relapsing-remitting or secondary-progressive) with disease activity (Steinman L, et al. N Engl J Med. 2022, 387(8):704-714). ULTIMATE I (N=549) and II (N=545) studies evaluated ublituximab 450 mg intravenous infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) or teriflunomide 14 mg oral once daily for 96 weeks in participants with RMS. Laboratory evaluations were conducted at Days 1 (predose), 2 (one day after initial infusion), 8, and 15 (predose); every 4 weeks at Weeks 4-24; every 12 weeks up to Week 96; and at Weeks 100 and 104. Cytopenias were evaluated both as laboratory abnormalities (grade based on cell counts) and treatment-emergent adverse events (TEAEs) based on medical judgment to be clinically significant (severity graded by investigator) according to the Common Terminology Criteria for Adverse Events, version 4.0. The safety population (pooled for both studies) included all participants who received at least one dose of study drug. The results are described in Figures 17A-17C.
(b) Results
[00535] In the pooled analysis, cytopenia as a TEAE (any grade/Grade >3) was reported in 25.7%/12.1% of ublituximab participants. The most commonly reported cytopenias (all Grade/Grade >3) were lymphopenia (18.7%/9.5%), neutropenia (3.3%/2.0%), leukopenia (3.3%/0.2%), and anemia (2.9%/0.0%). The median (quartile 1, quartile 3) time to resolution of Grade >3 lymphopenia TEAEs with ublituximab by the next assessment was 6.0 days (6.0, 6.0) and 13.0 days (7.0, 21.0) for neutropenia TEAEs. Excluding lymphopenia, rates of Grade >3 TEAEs were similar between ublituximab-treated participants (14.1%) and teriflunomide-treated participants (13.5%). In ublituximab-treated participants, across all study visits, cytopenias were more frequently reported as laboratory abnormalities than as TEAEs, and most of these laboratory abnormalities were Grade 1 or 2 events (Figure 17A). Decreased lymphocytes, leukocytes, hemoglobin, and neutrophils were reported in 90.8%, 38.1%, 23.6%, and 15.1% of ublituximab participants, respectively. Excluding Day 2, the proportion of participants with lymphopenia decreased from 90.8% to 32.2% (Figure 17B). On Day 2, 91% of participants had low lymphocyte counts (39.2% Grade >3); this dramatically reduced ~12 fold to only 7.8% of participants with low lymphocyte counts at Day 8 (<1% Grade >3), indicative of a transient postinfusion finding (Figure 17C). On Day 2, <1% of participants had low neutrophil counts; only 1 participant had one Grade 2 event and 16.6% of participants had low hemoglobin; all were Grade 1 or 2 events. After Day 2, lymphocyte counts below the lower limit of normal (<LLN) were observed at a low incidence for the remainder of the study. Of 102 participants with a TEAE of lymphopenia, 81 (79.4%) reported the event once on Day 2 and recovered by Day 8 without recurrence thereafter. Out of 52 participants with a Grade >3 lymphopenia or lymphocyte count decrease TEAE, only 2 participants experienced the TEAE outside of the Day 2 time point. Excluding lymphopenia, rates of Grade >3 TEAEs were 14.1% with ublituximab and 13.5% with teriflunomide.
[00536] Serious infections were not commonly associated with a decrease in lymphocyte or neutrophil counts <LLN. About 10.7% of serious infections occurred with lymphocytes <LLN within 1 month before or after onset of the infection, and 3.6% of serious infections occurred with neutrophils <LLN within 1 month before or after onset of the infection.
(c) Conclusion
[00537] Unique to the ULTIMATE I and II study designs was the assessment of laboratory values at Day 2, which revealed a transient decrease in lymphocytes that normalized in the majority of ublituximab-treated participants by Day 8. Excluding lymphopenia, the rate of Grade >3 TEAEs with ublituximab was comparable to teriflunomide.
8.18 Example 18 — Phase 3 Results of Ublituximab compared to Teriflunomide in Treating Patients with Relapsing Forms of Multiple Sclerosis (RMS) - Annualized Relapse Rate (ARR)
[00538] ULTIMATE I and ULTIMATE II, two identical, independent, phase 3 randomized, multi-center, double-blinded, active-controlled clinical trials, conducted in parallel, enrolled 1094 patients with relapsing forms of multple sclerosis (RMS) across 10 countries, to evaluate the safety and efficacy of the glycoengineered anti-CD20 antibody ubliximab versus teriflunomide in these patients.
[00539] Clinical evaluations were performed every 12 weeks, and CNS magnetic resonance imaging (MRI) assessments were performed at Weeks 12, 24, 48, and 96. The clinical use of MRI in the diagnosis of MS (Rovira, A. etal., Nat Rev Neurol 77:471-482 (2015)) and the assessment of MRI lesions as a surrogate for MS relapse (Sormani el al., Ann. Neurol. 65(3):268-275 (2009); Sormani et al., Lancet Neurol. 12.669-616 (2013)) is known. See also, Kraunzer, UW and Gauthier, SA, Ther Adv Neurol Disord. 70(6) 247-261 (2017).
[00540] The primary endpoint for the ULTIMATE I and ULTIMATE II phase 3 clinical studies was the Annualized Relapse Rate (ARR) following 96 weeks of treatment. ARR refers to the ratio of the sum of the subject’s RMS relapse counts divided by the sum of the subject’s treatment duration (in years). [00541] Key secondary endpoints for ULTIMATE I and ULTIMATE II included total number of gadolinium-enhancing T1 lesions on MRI by Week 96; total number of new and/or enlarging T2 hyperintense lesions by Week 96; and the proportion of subjects with No Evidence of Disease Activity (NED A) from Week 24 to Week 96.
[00542] In a pre-specified pooled analyis, key secondary endpoints included time to Confirmed Disability Progression (CDP) for at lesst 12 weeks. Tertiary analyes included time to Confirmed Disability Progression (CDP) for at least 24 weeks, time to Confirmed Disability Improvement (CDI) for at least 12 weeks; and time to Confirmed Disability Improvement (CDI) for at least 24 weeks.
[00543] These results will be provided in this and the following Examples and the associated Figures.
[00544] As mentioned above, the primary endpoint for the ULTIMATE I and ULTIMATE II phase 3 clinical studies was Annualized Relapse Rate (ARR) following 96 weeks of treatment. Both the ULTIMATE I and ULTIMATE II studies met their primary endpoint with ublituximab treatment demonstrating a statistically significant reduction in annualized relapse rate (ARR) over a 96- week period (p<0.005 in each trial). Ublituximab treatment resulted in an ARR of <0.10 in each of ULTIMATE I and ULTIMATE II.
[00545] More specifically, in ULTIMATE I, treatment with ublituximab (N =271) resulted in an ARR of 0.076 compared to 0.188 for teriflunomide (N=274), representing a relative reduction of 59.4% (p<0.0001). See, Figure 18A.
[00546] In ULTIMATE II, treatment with ublituximab (N = 272) resulted in an ARR of 0.091 compared to 0.178 for teriflunomide (N = 272), representing a relative reduction of 49.1% (p=0.0022). See, Figure 18B.
8.19 Example 19 — MRI Results: Reduction in Total Number of Gadolinium (Gd)-enhancing T1 Lesions with Ublituximab vs. Teriflunomide in the Phase 3 ULTIMATE I and II Studies of Patients with RMS
[00547] At 96 weeks, the total number of Gadolinium-enhancing (Gd+) lesions on MRI were reduced as a result of ublituximab treatment by 96.7% (N=265) and 96.5% (N=272), relative to treatment with teriflunomide (N=270; N=267) in ULTIMATE I and ULTIMATE II, respectively (pO.0001). See, Figure 19A (ULTIMATE I) and Figure 19B (ULTIMATE II). 8.20 Example 20 — MRI Results: Reduction in new or enlarging T2 lesions with Ublituximab vs. Terifhinomide in the Phase 3 ULTIMATE I and II Studies in Patients with RMS
[00548] At 96 weeks, the number of new or enlarging T2 lesions on MRI were reduced as a result of ublituximab treatment by 92.4% (N=265) and 90.0% (N=272), relative to treatment with teriflunomide (N=270; N=267) in ULTIMATE I and ULTIMATE II, respectively (pO.0001). See, Figure 20A (ULTIMATE I) and Figure 20B (ULTIMATE II).
8.21 Example 21 — Ublituximab is associated with Improved No Evidence of Disease Activity (NEDA) Status vs. Teriflunomide
[00549] In the ULTIMATE I study, 44.6% of ublituximab-treated patients (N=271) achieved no evidence of disease activiy (NEDA) status, compared to 15.0% of teriflunomide-treated patients (N=274), representing a 197% improvement over treatment with teriflunomide (p <0.0001). See, Figure 21A.
[00550] In the ULTIMATE II study, 43% of ublituximab-treated patients (N=272) achieved no evidence of disease activity (NEDA) status, compared to 11.4% of teriflunomide-treated patients (N=272), representing a 277% improvement over teriflunomide (p<0.0001). See, Figure 21B
8.22 Example 22 — Reduction in T1 Hypoin tense Lesions (“black holes”) With Ublituximab vs. Teriflunomide in the Phase 3 ULTIMATE I and II Studies in Patients with RMS
[00551] Background'. In this Example, the impact of ublituximab on T1 hypointense lesions on MRI in the ULTIMATE I and ULTIMATE II studies was evaluated. T1 “black holes” are hypointense lesions commonly seen on T1 -weighted images in patients with multiple sclerosis, and are indicative of chronic stage disease associated with white matter destruction, axonal loss, and irreversible clinical outcome. See, Kocsis, K. el al., Frontiers in Neurology 12'. 1-8 (March 2021).
[00552] Methods'. In the ULTIMATE I (N=549) and ULTIMATE II (N=545) phase 3 studies, the volume of T1 hypointense lesions was a tertiary endpoint in the individual studies. Pooled analyses of T1 hypointense lesions were performed post hoc. T1 hypointense lesion volume was assessed using mixed model repeated measures of the change in cubic root transformed volume from baseline at scheduled visits up to 96 weeks.
[00553] Results'. In the pooled analysis of ULTIMATE I and II, the mean change from baseline at 96 weeks in T1 hypointense lesion volume was 0.0101 mL for ublituximab-treated patients and 0.0491 mL for teriflunomide-treated patients, a difference of -0.0390 (95% confidence interval, -0.0585 to -0.0195; P<0.0001 for all post-baseline timepoints). The mean volume of T1 hypointense lesions (mL) at baseline, Week 24, Week 48, and Week 96 was 3.280, 3.266, 3.244, and 3.132 for ublituximab and 3.343, 3.360, 3.330, and 3.475 for teriflunomide, respectively. The mean number of new T1 hypointense lesions at 96 weeks was significantly reduced with ublituximab versus teriflunomide (1.5±3.55 vs 5.4±10.67; P<0.0001).
[00554] Percent (%) change reductions between ublituximab and teriflunomide for T1 lesion volume and T1 lesion number were 9.9% and 72.2%, respectively.
[00555] Conclusions'. In a pooled post hoc analyses in the ULTIMATE 1 and ULTIMATE II phase 3 clinical trials, significant reductions in both the volume and number of new T1 hypointense lesions were seen with ublituximab versus teriflunomide at 96 weeks.
8.23 Example 23 — Pharmacodynamics (PD) of B-Cell Depletion and
Pharmacokinetics (PK) of Ublituximab in Patients with RMS in ULTIMATE I and ULTIMATE II
[00556] In this Example, the pharmacodynamics (PD) of B-cell depletion and the pharmacokinetics (PK) of ublituximab treatment were evaluated in patients with RMS in the ULTIMATE I and ULTIMATE II phase 3 clinical studies.
[00557] Methods'. ULTIMATE I (N=549) and ULTIMATE II (N=545) evaluated ublituximab 450 mg intravenous 1-hour infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) or teriflunomide 14 mg oral once daily for 96 weeks in patients with RMS. The last dose of ublituximab was given at Week 72, with re-treatment starting on average (mean) 51-52 weeks later for patients enrolling in the open-label extension (OLE) study. PD and PK analyses were performed in the modified intention-to-treat population at pre-specified intervals. [00558] Results'. In a pooled post hoc analysis of ULTIMATE I and ULTIMATE II (N=543), the median number of CD 19+ B cells (cells/pL) at baseline in participants receiving ublituximab was 201.0. Starting at Week 1, Day 2, participants had a notable decrease from baseline in the median number of CD 19+ B cells (-193.0 [96.0% reduction]), which remained consistent through Week 104 (-183.0 [91.0% reduction]), 32 weeks after the last infusion. Prior to the first OLE infusion, median B-cell numbers had increased to 22.4% of baseline.
Pharmacokinetics and Absorption
Absorption
[00559] The pharmacokinetics (PK) of ublituximab following repeated IV infusions were adequately described by a two-compartment model with first-order elimination. Ublituximab exposures increased in a dose-proportional manner (i.e., linear pharmacokinetics) over the dose range of 150 to 600 mg in patients with RMS.
[00560] The administration of ublituximab by intravenous infusion resulted in a geometric mean steady-state AUC of 3000 mcg/mL per day (CV = 28%) and a mean maximum concentration of 139 mcg/mL (CV =15%).
Distribution
[00561] The population PK estimate of the central volume of distribution was 3.18 L.
Elimination
[00562] The mean terminal half life of ublituximab was estimated to be 22 days.
[00563] Conclusions'. In the ULTIMATE I and ULTIMATE II phase 3 clinical trials, peripheral B-cell numbers declined rapidly after the first ublituximab infusion and remained low during treatment, which may be consistent with ublituximab’ s mechanism of action.
8.24 Example 24 — Neutralizing Antibodies (NAbs) and Antidrug Antibodies (AD As) in the Ublituximab Phase 3 ULTIMATE I and II Studies in Patients with RMS
[00564] In this Example, the incidence of neutralizing antibodies (NAbs) and antidrug antibodies (AD As) against ublituximab in the ULTIMATE I and II studies were evaluated.
[00565] Methods'. The ULTIMATE I (N=549) and ULTIMATE II (N=545) studies evaluated ublituximab 450 mg intravenous infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) or teriflunomide 14 mg oral once daily for 96 weeks in patients with RMS. NAb and ADA analyses were performed in patients who received at least 1 ublituximab dose (safety population) and were evaluable for NAbs and AD As; ULTIMATE I and II data were pooled (N=534).
[00566] Results'. The proportion of patients receiving ublituximab testing positive for NAbs and AD As was 2.4% and 17.8% at baseline and 6.4% and 86.5% postbaseline, respectively. Patients testing positive at baseline did not necessarily test positive at postbaseline timepoints. Treatment-emergent (TE)-NAbs were present in 4.3%, 3.4%, 1.1%, and 1.1% of patients at Weeks 24, 48, 72, and 96, respectively. The proportion of patients with TE-ADAs was 48.9%, 69.1%, 48.1%, 28.1%, and 16.5% at Weeks 3, 24, 48, 72, and 96, respectively. Annualized relapse rates (ARRs) were 0.03 in NAb-positive (postbaseline) (n=34), 0.11 in NAb-negative (postbaseline) (n=500), 0.10 in TE- ADA-positive (n=434), and 0.12 in TE- ADA-negative (n=100) patients; patient numbers were small for the NAb-positive group. All Grade/Grade >3 infusion-related reactions (IRRs) occurred in 48.4%/3.0% of TE-ADA-positive and in 42.0%/2.0% of TE- ADA-negative patients. IRR AEs (all Grade) that occurred with >2% higher frequency in TE-ADA-positive vs TE- ADA-negative patients were pyrexia (10.1% vs 7.0%), chills (8.3% vs 6.0%), nausea (3.7% vs 1.0%), and lymphocyte decreased (3.2% vs 1.0%). Patient postbaseline NAb and TE-ADA status did not appear to be associated with particular baseline demographic or disease characteristics.
[00567] Conclusions'. In the Phase 3 ublituximab studies, a low incidence of TE-NAbs was observed, and the majority of patients developed TE-ADAs. The proportion of patients with TE- NAbs and TE-ADAs declined after 24 weeks.
8.25 Example 25 — Pre-specified Pooled Disability Results - Confirmed Disability Progression (CDP) and Confirmed Disability Improvement (CDI) in the Ublituximab Phase 3 ULTIMATE I and II Studies in Patients with RMS
[00568] CDP'. As used herein, CDP refers to the proportion of patients (%) to reach the outcome of CDP for at least 12 weeks (CDP 12) or for 24 weeks (CDP 24) from baseline to week 96 while on ublituximab or teriflunomide treatment. Additionally, CDP refers to the time to reach the outcome of CDP for at least 12 weeks (CDP 12) or for 24 weeks (CDP 24) from baseline to week 96 while on ublituximab or teriflunomide treatment. See, Ontaneda, D. et al., Lancet 359:1357-1366 (2017). [00569] A very low rate of disability progression was observed across all treatment groups (i.e., ublituximab and teriflunomide). Only 5.2% of ublituximab-treated RMS patients showed a 12- week time to onset of Confirmed Disability Progression (CDP), compared to 5.9% of RMS patients treated with teriflunomide. See, Figure 22A. Only 3.3% of ublituximab-treated RMS patients showed a 24-week time to onset of CDP, compared to 4.8% of RMS patients treated with teriflunomide, See, Figure 22B. Neither result was statistically different.
[00570] CDI: As used herein, CDI refers to the proportion of patients (%) to reach the outcome of CDI for at least 12 weeks (CDI 12) or for at least 24 weeks (CDI 24) from baseline to week 96 while on ublituximab or teriflunomide treatment. Additionally, CDI refers to the time to reach the outcome of CDI for at least 12 weeks (CDI 12) or for 24 weeks (CDI 24) from baseline to week 96 while on ublituximab or teriflunomide treatment. See, Cree, B. el al., Mult. Scler. 27(14):2219-2231 (2021).
[00571] The proportion of participants who achieved CDI for at least 12 weeks was higher in the ublituximab group (12.0%) compared with the teriflunomide group (6.0%). The difference between treatment groups for time to CDI for at least 12 weeks was statistically significant in favor of ublituximab using a stratified log-rank test (116% increase; p=0.0003]).
[00572] The proportion of participants who achieved CDI for at least 24 weeks was higher in the ublituximab group (9.6%) compared with the teriflunomide group (5.1%). The difference between treatment groups for time to CDI for at least 24 weeks was statistically significant in favor of ublituximab using a stratified log-rank test (103% increase; p=0.0026). See, Figure 23A for 12- week CDI analysis and Figure 23B for 24-week CDI analysis.
8.26 Example 26 — Ublituximab is Associated with Significant Improvement in the Multiple Sclerosis Functional Composite (MSFC) Score vs. Teriflunomide: Results from the Phase 3 ULTIMATE I and ULTIMATE II Studies
[00573] Improvements in disability were observed with ublituximab in both ULTIMATE I and ULTIMATE II studies, with significantly more patients achieving 12- week or 24-week Confirmed Disability Improvement (CDI) versus teriflunomide. See, Example 25.
[00574] In the present Example, the effects of ublituximab on the Multiple Sclerosis Functional Composite (MSFC) score and its components were studied compared to teriflunomide. The components of MSFC included: a 9-Hole Peg Test (9-HPT), to assess arm and hand function; a Timed 25-Foot Walk (T25FW) to assess leg function; and a Paced Auditory Serial Additional Test (PASAT) to assess cognitive function. Fischer, JS et al., Mult. Scler. 5:244-250 (1999).
[00575] Design/Methods: In the ULTIMATE I and ULTIMATE II studies, a total of 1094 RMS patients from 10 countries were studied, and received ublituximab 450 mg IV infusion every 24 weeks (following day 1 infusion of 150 mg) or teriflunomide 14 mg oral once-daily for 96 weeks. The MSFC (a tertiary endpoint of ULTIMATE 1 and II) was administered at baseline and every 12 weeks thereafter. Both the MSFC and components (9-HPT, PASAT, T25FW; post hoc analyses) were pre-specified endpoints and were analyzed using the modified intention-to- treat (ITT) population. A change from baseline in MSFC and components were analyzed using a mixed model repeated measures of change from baseline at all post-baseline timepoints. The model included treatment, region, baseline EDSS strata, visit, treatment-by-visit interaction, and baseline value as covariates, and used an unstructured covariance matrix, restricted maximum likelihood estimation, and the Satterthwaite method for degrees.
[00576] Results: Ublituximab treatment in patients with RMS was associated with significant improvement in the overall Multiple Sclerosis Functional Composite (MSFC) score from baseline as compared to patients treated with teriflunomide in both ULTIMATE I and ULTIMATE II (p=0.0484 and p=0.0171), respectively.
[00577] In ULTIMATE I, patients administered ublituximab (N=271) achieved a 76.4% improvement in MSFC score compared to patients taking teriflunomide (N=274). See Figure 24A. In ULTIMATE II, patients administered ublituximab (N=272) achieved an 89.6% improvement in MSFC score compared to patients taking teriflunomide (N=272). See Figure 24B).
[00578] In both ULTIMATE I and ULTIMATE II, ublituximab provided significant improvements from baseline to 96 weeks in mean timed 25 foot walk (T25FW) score versus teriflunomide. See, Figure 25A (ULTIMATE I) and Figure 25B (ULTIMATE II). T25FW was statistically significant in ULTIMATE II, but not in ULTIMATE I (p=0.0446 and p=0.2861, respectively).
[00579] In both ULTIMATE I and ULTIMATE II, the change in mean Paced Auditory Serial Additional Test (PASAT) score from baseline to 96 weeks was similar between ublituximab and teriflunomide, as shown in Figure 26A (ULTIMATE I) and Figure 26B (ULTIMATE II). The PAS AT scores were not statistically significant in either ULTIMATE I or II (p=0.7444 and p=0.7915), respectively.
[00580] In both ULTIMATE I and ULTIMATE II, ublituximab provided a significant increase in mean 9-Hole Peg Test (9-HPT) scores from baseline to 96 weeks versus teriflunomide. The 9-HPT was statistically significant in both ULTIMATE I and II (p=0.0084 and p=0.0106, respectively). See, Figure 27A (ULTIMATE I) and Figure 27B (ULTIMATE II). [00581] Conclusions: Ublituximab treatment was associated with significant improvement in multiple sclerosis functional composite (MSEC) score versus teriflunomide in both the ULTIMATE I and ULTIMATE II phase 3 studies. The improved MSEC score was driven by improvements in disability, as measured by the 9-Hole Peg Test (9-HPT) and Timed 25-Foot Walk (T25FW).
8.27 Example 27 — Percent Brain Volume Change (PBVC) on MRI
[00582] A post-hoc analysis of the difference in brain volume between Week 24 and Week 96 in ULTIMATE I showed that there was no difference in the brain volume decline, as measured by mean PBVC, between the ublituximab and teriflunomide arms. See, Figure 28A.
[00583] In ULTIMATE II, the brain volume decline, as measured by mean PBVC, was lower for the ublituximab arm than the teriflunomide arm. See, Figure 28B.
8.28 Example 28 — Infusion-Related Reactions (IRRs) with Ublituximab in Phase 3 ULTIMATE I and ULTIMATE II studies
[00584] In post-hoc analyses from the Phase 3 ULTIMATE I and ULTIMATE II studies, pooled IRR rates from both trials were analyzed and characterized by time couse and severity. [00585] Methods'. ULTIMATE I (N=549) and II (N=545) evaluated ublituximab 450 mg intravenous 1-hour infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) or teriflunomide 14 mg oral once daily for 96 weeks in patients with RMS. The teriflunomide group received placebo infusions; the ublituximab group received oral placebo. Participants received antihistamine and corticosteroid premedication 30-60 minutes prior to ublituximab/placebo infusions. Pooled IRR data from both trials were analyzed. [00586] Results'. Across ULTIMATE studies, the total number of infusions was 2644 (ublituximab) and 2637 (placebo). IRR rates were 47.7% (ublituximab) and 12.2% (placebo). 43.3% of ublituximab-treated patients had an IRR at the first infusion. 68.8% (n=181) had 1 IRR only; of these, 90.6% experienced the IRR at first dose. In patients experiencing >1 IRR (n=82), 87.8% experienced the first IRR during the first dose; thus, among patients with a first-dose IRR, 69.5% did not have an IRR recurrence. Pyrexia was the most common IRR (9.5% of ublituximab-treated patients). One patient experienced a Grade 4 IRR (anaphylaxis) with ublituximab at the Day 15 infusion (drug interrupted/withdrawn) following two Grade 1 IRRs at the first infusion (both reported as influenza-like syndrome); all IRRs resolved. Overall, 96.6% of total ublituximab infusions were completed without interruption.
[00587] Conclusions'. IRRs, with pyrexia being the most common, were the prevailing adverse event with ublituximab in the ULTIMATE I and II phase 3 trials. The majority of IRRs occurred at the first dose and had minimal impact on infusion completion.
8.29 Example 29 — Ublituximab Reduces Thalamic Volume Loss and New Lesion Formation in Participants of the ULTIMATE I & II Phase 3 Studies
[00588] Thalamic integrity, a measure of deep grey matter, is affected by damage in tracts projecting into and out of the thalamus. The impact of ublituximab on thalamic volume and on new lesion formation was evaluated in the ULTIMATE I and II studies during and after Year 1. [00589] Methods: The placebo-controlled ULTIMATE I (N=549) and II (N=545) studies evaluated ublituximab 450 mg intravenous infusion every 24 weeks (following Day 1 infusion of 150 mg and Day 15 infusion of 450 mg) vs. teriflunomide 14 mg orally once daily for 96 weeks in participants with RMS. In the pooled modified intention-to-treat population, the percentage thalamic volume change was measured from baseline to Week 96 and in yearly epochs using paired Jacobian integration. Post hoc analyses of changes over both years and at yearly intervals were performed for T1 hypointense lesion volume, new T1 hypointense lesion counts, and new/enlarging T2 lesion counts.
[00590] Results: Ublituximab treatment significantly reduced thalamic volume loss relative to teriflunomide by 22% over 2 years. The least squares (LS) mean percentage change from baseline to Week 96 was -1.34 (95% CI: -1.56, -1.12) vs -1.71 (95% CI: -1.93, -1.50) for ublituximab vs teriflunomide, respectively (P=0.0013). For T1 hypointense lesion volume (mL), LS mean percent change from baseline to Week 96 for ublituximab was 6.32 (95% CI: -0.82, 13.46) vs 24.87 (95% CI: 17.77, 31.96) for terifhrnomide (P<0.0001). The reduction in T1 hypointense lesion volume in Year 2 of treatment was more pronounced than in Year 1, with the same pattern seen for T1 hypointense lesion count and new/enlarging T2 lesions.
[00591] Conclusion: Ublituximab reduced thalamic volume loss and T1 hypointense lesion volume (markers of brain tissue loss) compared with teriflunomide. New T1 and T2 lesion formation was almost totally suppressed after the first year of treatment.
[00592] Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A composition comprising a population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody in the population is expressed from one or more nucleic acid sequences encoding a heavy chain comprising the amino acid sequence of SEQ ID NO: 1 and a light chain comprising the amino acid sequence of SEQ ID NO:2, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising about 20 to 40% fucosylated glycans and optionally about 10 to 20% galactosylated glycans.
2. The composition of claim 1 , wherein: the N-glycan profile comprises 23% to 36% fucosylated glycans, optionally about 30% fucosylated glycans; and/or the N-glycan profile comprises 16% to 18% galactosylated glycans, optionally about 17% galactosylated glycans.
3. The composition of claim 1 or 2, wherein: the relative abundance of fucosylated glycans is the percent of fucosylated glycans among all glycans in the N-glycan profile; and/or the relative abundance of galactosylated glycans is the percent of galactosylated glycans among all glycans in the N-glycan profile.
4. The composition of any one of claims 1-3, wherein the N-glycan profile comprises 12% to 30% bisecting N-glycans, optionally about 18% bisecting N-glycans.
5. The composition of claim 4, wherein the bisecting N-glycans comprise one or more of GOB, G0FB, G1FB, G2FBS1, and G2FBS2.
6. The composition of any one of claims 1-5, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising less than 5% sialylated glycans, optionally wherein the N-glycan profile comprises less than 4%, 3%, 2.5%, 2%, 1%, or 0.5% sialylated glycan, optionally wherein the N-glycan profile comprises no detectable amount of sialylated glycan.
7. The composition of any one of claims 1 -6, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising 0.1% to 1.5% Man5 N-glycan, optionally wherein the N-glycan profile comprises 0.4% to 0.7% Man5 N-glycan, optionally wherein the N-glycan profile comprises about 0.6% Man5 N-glycan, optionally wherein Man5 N-glycan is the only high mannose species in the N-glycan profile.
8. The composition of any one of claims 1-7, wherein the population of anti-CD20 antibody proteins comprises 0.20 to 0.40 mol isoaspartate per mol protein, optionally 0.25 to 0.35 mol isoaspartate per mol protein.
9. The composition of any one of claims 1-8, wherein glutamate at position 1 of the heavy chain is a pyroglutamate and glutamate at position 1 of the light chain is a pyroglutamate.
10. The composition of any one of claims 1-9, wherein the population of anti-CD20 antibody proteins has an N-glycan profile comprising a relative abundance ratio of 0.1 to 0.15 G1 to GO N-glycans and/or a relative abundance ratio of 0.5 to 0.9 GIF to G1 N-glycans.
11. The composition of any one of claims 1-10, wherein the population of anti-CD20 antibody proteins further comprises at least two N-glycans within the following relative abundance ranges:
(a) 0.3% to 2% G0-GN;
(b) 0.1% to 2% G0F-GN;
(c) 30% to 60% GO;
(d) 0.1% to 1% G1-GN;
(e) 5% to 20% GOB;
(f) 5% to 30% G0F;
(g) 0.1% to 1.5% Man5; (h) l% to 15% G0FB;
(i) l% to 13% Gl;
(j) 0.5% to 10% Gl’;
(k) 0.5% to 6% GIB;
(l) 0.5% to 12% GIF;
(m) 0.1% to 3% GIF’;
(n) 0.1% to 3% G1FB;
(o) 0.1% to 2% G2; and
(p) 0.1% to 2% G2F. optionally wherein the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
(a) 0.8% to 1.1% GO-GN;
(b) 0.5% to 1.1% G0F-GN;
(c) 42.5% to 48.8% GO;
(d) 0.3% to 0.6% Gl-GN;
(e) 9.5% to 14.1% GOB;
(f) 12.8% to 19.7% G0F;
(g) 0.4% to 0.7% Man5;
(h) 5.1% to 7.0% G0FB;
(i) 5.7% to 6.4% Gl;
(j) 2.7% to 3.3% Gl’;
(k) 1.4% to 2.0% GIB;
(l) 2.6% to 4.2% GIF;
(m) 1.1% to 1.6% GIF’;
(n) 1.1% to 1.8% G1FB;
(o) 0.5% to 0.7% G2; and
(p) 0.3% to 0.5% G2F, optionally wherein the population of anti-CD20 antibody proteins further comprises at least two N-glycans in the following relative abundance ranges:
(a) 0.9% G0-GN;
(b) 0.8% G0F-GN; (c) 46.1% GO;
(d) 0.5% Gl-GN;
(e) 10.9% GOB;
(f) 17.0% G0F;
(g) 0.6% Man5;
(h) 6.0% G0FB;
(i) 6.1% Gl;
(j) 2.9% Gl’;
(k) 1.6% GIB;
(l) 3.2% GIF;
(m) 1.3% GIF’;
(n) 1.3 G1FB;
(o) 0.5% G2; and
(p) 0.3% G2F.
12. The composition of claim 11, wherein the population of anti-CD20 antibody proteins further comprises at least three, four or five N-glycans within the following relative abundance ranges:
(a) 0.3% to 2% GO-GN;
(b) 0.1% to 2% GOF-GN;
(c) 30% to 60% GO;
(d) 0.1% to 1% G1-GN;
(e) 5% to 20% GOB;
(f) 5% to 30% G0F;
(g) 0.1% to 1.5% Man5;
(h) l% to 15% G0FB;
(i) l% to 13% Gl;
(j) 0.5% to 10% Gl’;
(k) 0.5% to 6% GIB;
(l) 0.5% to 12% GIF;
(m) 0.1% to 3% GIF’; (n) 0.1% to 3% G1FB;
(o) 0.1% to 2% G2; and
(p) 0.1% to 2% G2F.
13. The composition of any one of claims 1-12, wherein the N-glycan profile of the population of anti-CD20 antibody proteins is determined using a method comprising:
(a) incubating the population of anti-CD20 antibody proteins with an enzyme, wherein the enzyme catalyzes releasing of the N-glycans from the anti-CD20 antibody;
(b) measuring the relevant abundance of the released N-gylcans using one or more methods selected from chromatography, mass spectrometry, capillary electrophoresis, and the combination thereof, optionally wherein the method further comprises after step (a) and before step (b) the following steps:
(c) purifying the N-glycans; and
(d) labeling the N-glycans with a fluorescent compound, optionally wherein the enzyme is PNGase F and/or the fluorescent compound is 2- aminobenzamide (2-AB).
14. The composition of any one of claims 1-13, wherein less than 10% of the anti- CD20 antibody proteins in the population is non-glycosylated, optionally wherein less than 5% of the anti-CD20 antibody proteins in the population is non-glycosylated, optionally wherein less than 1% of the anti-CD20 antibody proteins in the population is non-glycosylated.
15. The composition of any one of claims 1-14, wherein the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) 8.0% to 10.0% a-helix;
(b) 32.0% to 36.0% Anti-parallel P-sheet;
(c) 5.0% to 6.0% Parallel P-sheet; (d) 16.0% to 18.0% P-Turn; and
(e) 35.0% to 36.0% random coil, optionally wherein the population of anti-CD20 antibody proteins comprises secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) 8.0% to 10.0% a-helix;
(b) 32.0% to 36.0% Anti-parallel P-sheet;
(c) 5.0% to 6.0% Parallel P-sheet;
(d) 16.0% to 18.0% P-Turn; and
(e) 35.0% to 36.0% random coil, optionally wherein the population of anti-CD20 antibody proteins comprises two or more secondary structures as determined by circular dichroism at 205 nm to 260 nm as follows:
(a) about 9.0% a-helix;
(b) about 33.0% Anti-parallel P-sheet;
(c) about 5.6% Parallel P-sheet;
(d) about 17.5% P-Turn; and
(e) about 35.2% random coil.
16. The composition of any one of claims 1-15, wherein the population of anti-CD20 antibody proteins further comprises one or more of the following post-translational modifications at the specified abundance:
Figure imgf000184_0001
Figure imgf000185_0001
optionally wherein the one or more of the post-translational modifications are measured by peptide mapping using liquid chromatography-mass spectrometry (LC-MS).
17. The composition of any one of claims 1-16, wherein the population has an amount of total protein of 25.5-25.8 mg/mL as measured by absorbance at 280 nm.
18. The composition of any one of claims 1-17, wherein the anti-CD20 antibody proteins in the population induces greater cytotoxicity in a cell-based antibody-dependent cellular cytotoxicity (ADCC) assay compared to obinutuzumab, ofatumumab, rituximab, veltuzumab, ibritumomab tiuxetan and/or ocrelizumab.
19. The composition of any one of claims 1-18, wherein the population has: a relative potency of 90 to 163% in a cell-based ADCC assay compared to a commercial reference standard; a relative potency of 78% to 116% or 73% to 128% in a cell-based complement dependent cytotoxicity (CDC) assay compared to that of a commercial reference standard; a relative potency of 92 to 118% or 82 to 138% in a cell-based CD20 binding activity bioassay compared to that of a commercial reference standard; a KD value 30 to 70 nM in an FcyRIIIa- 158V binding assay as measured by surface plasmon resonance; a KD value 500 to 1000 nM in an FcyRIIIa 158F binding assay as measured by surface plasmon resonance; significantly higher binding affinity to FcyRIIIa 158V or FcyRIIIa 158F than rituximab; a relative potency of 88 to 113% or 86 to 116% in a Clq binding assay as measured by ELISA compared to a commercial reference standard; and/or a relative potency of 106 to 126% in a CD 16 activity assay compared to a commercial reference standard.
20. The composition of any one of claims 1-19, wherein the population has: 99.2 to 99.9% monomers as detected by size exclusion chromatography (SEC); 0.1 to 0.8% dimers as detected by SEC; undetectable level of aggregates as detected by SEC; and/or undetectable level of fragments as detected by SEC.
21. The composition of any one of claims 1-20, wherein the population has: 93.6 to 95.9% IgG after purification by non-reduced capillary gel electrophoresis (CGE); 0.1 to 0.3% high molecular weight species (HMWS) after purification by non-reduced CGE; 0.7 to 1.2% free light chain (LC) after purification by non-reduced CGE; and/or 97.7 to 98.0% heavy chain plus light chain species (HC + LC) after purification by reduced CGE.
22. The composition of any one of claims 1-21, wherein the population has: 20 to 25% acidic isoforms as detected by imaged capillary isoelectric focusing (iCIEF); 50 to 60% main isoforms as detected by iCIEF; 20 to 30% basic isoforms as detected by iCIEF; and/or an average molar ratio of free thiol to anti-CD20 antibody of about 2.0 to 2.2.
23. The composition of any one of claims 1-22, wherein: the amino acid sequence of the anti-CD20 antibody in the population comprises a deletion of the N-terminal residue, optionally a deletion of up to 5 N-terminal residues, optionally a deletion of up to 10 N-terminal residues; and/or terminal lysine amino acid residue of the heavy chain in the anti-CD20 antibody in the population is truncated.
24. A pharmaceutical formulation comprising the composition of any one of claims 1 - 23, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 10 mg/mL to 50 mg/mL, optionally wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL.
25. The pharmaceutical formulation of claim 24, further comprising one or more of the following: sodium chloride, trisodium citrate dehydrate, polysorbate 80, and hydrochloric acid.
26. The pharmaceutical formulation of claim 25, comprising about 9.0 mg/mL of sodium chloride, about 7.4 mg/mL of trisodium citrate dehydrate, about 0.7 mg/mL of polysorbate 80, and/or about 0.4 mg/mL of hydrochloric acid.
27. The pharmaceutical formulation of any one of claims 24-26, wherein the anti- CD20 antibody is present in a single dosage form.
28. A pharmaceutical formulation comprising:
(i) the composition of any one of claims 1-23, wherein the composition comprises a single dosage form of the population of anti-CD20 antibody proteins, wherein the anti-CD20 antibody is present in the pharmaceutical formulation at a concentration of about 25 mg/mL,
(ii) about 9.0 mg/mL of sodium chloride,
(iii) about 7.4 mg/mL of trisodium citrate dehydrate,
(iv) about 0.7 mg/mL of polysorbate 80, and
(v) about 0.4 mg/mL of hydrochloric acid.
29. A single batch preparation of the population of anti-CD20 antibody proteins as recited in the method of any one of claims 1 -23 or the pharmaceutical formulation of any one of claims 24-28, wherein the single batch comprises at least 100 g, at least 120 g, or at least 150 g of the anti-CD20 antibody proteins.
30. A population of anti-CD20 antibody proteins as recited in the method of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28, produced in a 15,000 L or 20,000 L bioreactor.
31. A method of treating multiple sclerosis (MS) in a subject in need thereof, wherein the method comprises administering to the subject a therapeutically effective amount of the composition of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28.
32. The method of claim 31, wherein the MS is a relapsing form of MS (RMS), optionally wherein the RMS is selected from clinically isolated syndrome (CIS), relapsing-remitting MS (RRMS) and active secondary progressive MS (SPMS).
33. The method of claim 31 or 32, wherein the composition or the pharmaceutical formulation is administered as i) a first infusion at a dose of about 150 mg of the the anti-CD20 antibody protein, ii) a second infusion two weeks later at a dose of about 450 mg of the the anti- CD20 antibody protein, and iii) subsequent infusions every 24 weeks or six months at a dose of about 450 mg of the the anti-CD20 antibody protein.
34. The method of any one of claims 31-33, wherein administration of the composition or the pharmaceutical formulation to the subject results in one or more of the following pharmacokinetic parameters:
(a) an AUC between 2,160 pg/mL and 3,840 pg/mL;
(b) a Cmax between 118,011 ng/mL and 159,989 ng/mL;
(c) a Cmin between 40 ng/mL and 375 ng/mL; and
(d) a Cavg is between 6,437 ng/mL and 11,443 ng/mL, optionally wherein administration of the composition or the pharmaceutical formulation to the subject results in one or more of the following pharmacokinetic parameters: (a) an AUC about 3,000 pg/mL;
(b) a Cmax about 139,000 ng/mL;
(c) a Cmin about 139 ng/mL; and
(d) a Cavg about 8,940 ng/mL.
35. The method of any one of claims 31-34, wherein the method comprises a treatment period of at least 96 weeks.
36. The method of any one of claims 31-35, wherein the subject has been premedicated with a corticosteroid 30-60 minutes prior to administration of the composition or the pharmaceutical formulation, optionally wherein the corticosteroid is methylprednisone or dexamethasone, optionally wherein the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
37. The method of any one of claims 31-36, wherein the subject has been premedicated with an antihistamine 30-60 minutes prior to administration of the composition or the pharmaceutical formulation, optionally wherein, the antihistamine is diphenhydramine HC1, optionally wherein the diphenhydramine HC1 is administered at a dose of about 25-50 mg.
38. The method of any one of claims 31-37, wherein the subject has been premedicated with an antipyretic 30-60 minutes prior to administration of the composition or the pharmaceutical formulation, optionally wherein the antipyretic is acetaminophen or an antipyretic bioequivalent thereto.
39. The method of any one of claims 31-38, wherein the subject has an Expanded Disability Status Scale (EDSS) score of from 0 to 5.5 prior to treatment.
40. The method of any one of claims 31-39, wherein the subject is diagnosed with RMS in accordance to McDonald Criteria (2010).
41. A method of treating multiple sclerosis (MS) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the composition of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28, wherein administration of the composition or the pharmaceutical formulation results in no evidence of disease activity (NED A) in the subject 24-96 weeks after the administration.
42. The method of claim 41, wherein administration of the composition or the pharmaceutical formulation results in NED A in the subject 24 weeks after the administration.
43. A method of treating multiple sclerosis (MS) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the composition of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28, wherein administration of the composition or the pharmaceutical formulation results in a transient decrease in lymphocyte count in the subject.
44. The method of claim 43, wherein the lymphocyte count is normalized by day 8 of the administration.
45. The method of any one of claims 41-44, wherein the MS is a relapsing form of MS (RMS).
46. A method of reducing annualized relapse rate (ARR) in a subject with relapsing forms of multiple sclerosis (MS), comprising administering to the subject an effective amount of the composition of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28, the method comprising: administering an intravenous infusion of the composition or the pharmaceutical formulation in a multi-infusion dosage regimen, the dosage regimen comprising: a) a first infusion comprising 150 mg of the anti-CD20 antibody protein at day 1 ; b) a second infusion comprising 450 mg of the anti-CD20 antibody protein at about 2 weeks after the first infusion; c) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the first infusion; and d) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the prior infusion,.
47. The method of claim 46, wherein the effective amount of the composition or the pharmaceutical formulation is sufficient to result in an ARR of 0.091 or an ARR of 0.076.
48. The method of claim 46 or 47, wherein duration of the second infusion, the first subsequent infusion, and the one or more subsequent infusions of the anti-CD20 antibody protein is about one hour.
49. A method of treating relapsing forms of multiple sclerosis (MS) in a subject in need thereof, comprising administering to the subject an effective amount of the composition of any one of claims 1-23 or the pharmaceutical formulation of any one of claims 24-28, the method comprising: administering an intravenous infusion of the composition or the pharmaceutical formulation in a multi-infusion dosage regimen, the dosage regimen comprising: e) a first infusion comprising 150 mg of the anti-CD20 antibody protein at day 1; f) a second infusion comprising 450 mg of the anti-CD20 antibody protein at about 2 weeks after the first infusion; g) a first subsequent infusion comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the first infusion; and h) one or more subsequent infusions comprising 450 mg of the anti-CD20 antibody protein at about 24 weeks or at about six months from the prior infusion, wherein duration of the second infusion, the first subsequent infusion, and the one or more subsequent infusions of the anti-CD20 antibody protein is about one hour.
50. The method of any one of claims 41-49, further comprising pre-medicating the subject with a corticosteroid and an antihistamine, 30-60 minutes prior to administration of the composition or the pharmaceutical formulation.
51. The method of claim 50, wherein the corticosteroid is methylprednisone or dexamethasone, optionally wherein the methylprednisone is administered at a dose of about 100 mg and/or the dexamethasone is administered at a dose of about 10-20 mg.
52. The method of any one of claims 46-51, wherein the intravenous infusion of the composition or the pharmaceutical formulation is prepared in 250 mL of 0.9% Sodium Chloride Injection.
53. The method of any one of claims 46-52, wherein the first subsequent infusion is at about 24 weeks from the first infusion.
54. The method of any one of claims 46-53, wherein the one or more subsequent infusions is at about 24 weeks from the prior infusion.
55. The method of any one of claims 46-54, wherein the first subsequent infusion is at about 6 months from the first infusion.
56. The method of any one of claims 46-55, wherein the one or more subsequent infusions is at about 6 months from the prior infusion.
57. The method of any one of claims 46-56, wherein duration of the first infusion of the anti-CD20 antibody protein is about four hours, optionally wherein the first infusion of the anti-CD20 antibody protein is infused at a rate of 10 mL per hour for the first 30 minutes; 20 mL per hour for the next 30 minutes; 35 mL per hour for the next hour; and 100 mL per hour for the remaining two hours.
58. The method of any one of claims 48-57, wherein the second infusion, the first subsequent infusion, and the one or more subsequent infusion of the anti-CD20 antibody protein is infused at a rate of 100 mL per hour for the first 30 minutes, and 400 mL for the remaining 30 minutes.
59. The method of any one of claims 46-58, wherein the multi-infusion dosage regimen of the anti-CD20 antibody protein alleviates or delays progression of the symptoms of MS.
60. The method of any one of claims 46-59, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of gadolinium-enhancing T1 lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
61. The method of any one of claims 46-60, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a reduced total number of new and enlarging T2 hyperintense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
62. The method of any one of claims 46-61, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased no evidence of disease activity (NED A) status, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
63. The method of any one of claims 46-62, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves increased Confirmed Disability Improvement (CDI), as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
64. The method of any one of claims 46-63, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an increased Multiple Sclerosis Functional Composite (MSFC) score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
65. The method of any one of claims 46-64, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved timed 25-Foot Walk (T25FW) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
66. The method of any one of claims 46-65, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves an improved 9-Hole Peg test (9-HPT) Score, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
67. The method of any one of claims 46-66, wherein the subject administered the multi-infusion dosage regimen of the anti-CD20 antibody protein achieves a significant reduction in both volume and number of new T1 hypointense lesions per MRI scan, as compared to a subject that received orally administered 14 mg of teriflunomide daily during the same treatment period.
68. The method of any one of claims 46-67, wherein the multi-infusion dosage regimen of the anti-CD20 antibody protein results in a geometric mean steady-state AUC of 3000 mcg/mL per day (CV = 28%) and a mean maximum concentration of 139 mcg/mL (CV =15%).
69. The method of any one of claims 45-68, wherein the RMS comprises a clinically isolated syndrome ("CIS"); relapsing-remitting MS ("RRMS"); or active secondary progressive MS ("SPMS").
70. The method of any one of claims 45-69, wherein the subject is diagnosed with RMS in accordance to McDonald Criteria (2010).
71. The method of any one of claims 31 -70, wherein the subject is a human.
72. A method for inactivating a virus or adventitious agents in rat myeloma cells expressing the anti-CD20 antibody proteins recited in the composition of any one of claims 1-23, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
73. A method for reducing immunogenicity of the anti-CD20 antibody proteins recited in the composition of any one of claims 1-23, wherein the method maintains suitability for antibody production in a 15,000 L or 20,000 L bioreactor.
PCT/US2023/067711 2022-06-01 2023-05-31 Anti-cd20 antibody compositions WO2023235764A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263347852P 2022-06-01 2022-06-01
US63/347,852 2022-06-01
US202263421078P 2022-10-31 2022-10-31
US63/421,078 2022-10-31
US202363445082P 2023-02-13 2023-02-13
US63/445,082 2023-02-13

Publications (1)

Publication Number Publication Date
WO2023235764A1 true WO2023235764A1 (en) 2023-12-07

Family

ID=89025759

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/067711 WO2023235764A1 (en) 2022-06-01 2023-05-31 Anti-cd20 antibody compositions

Country Status (1)

Country Link
WO (1) WO2023235764A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1985633A1 (en) * 2007-04-26 2008-10-29 LFB Biotechnologies Kit of parts for the treatment of cancer or infectious diseases
US20200323981A1 (en) * 2012-11-02 2020-10-15 Tg Therapeutics, Inc. Combination of Anti-CD20 Antibody and PI3 Kinase Selective Inhibitor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1985633A1 (en) * 2007-04-26 2008-10-29 LFB Biotechnologies Kit of parts for the treatment of cancer or infectious diseases
US20200323981A1 (en) * 2012-11-02 2020-10-15 Tg Therapeutics, Inc. Combination of Anti-CD20 Antibody and PI3 Kinase Selective Inhibitor

Similar Documents

Publication Publication Date Title
EP3310378B1 (en) Anti-cll-1 antibodies and methods of use
EP3310814B1 (en) Humanized and affinity matured antibodies to fcrh5 and methods of use
JP2023531705A (en) Anti-CCR8 antibody therapy: biomarkers and combination therapy
TWI335821B (en) Immunoglobulin variants and uses thereof
JP6055615B2 (en) DACHYP Compositions and Methods
EP3374399A1 (en) Composition and methods for anti-tnfr2 antibodies
TW201938200A (en) Anti-MCT1 antibodies and uses thereof
EP2463368B1 (en) Humanized anti-amyloid-b oligomer antibody
US20230049152A1 (en) Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
CA3145940A1 (en) Anti-cd39 antibody compositions and methods
WO2023235764A1 (en) Anti-cd20 antibody compositions
EP4286010A1 (en) Anti-cd20 antibody compositions
EP4286414A1 (en) Anti-cd20 antibody compositions
EP4286413A1 (en) Anti-cd20 antibody compositions
TW202413404A (en) Anti-cd20 antibody compositions
US11965032B1 (en) Anti-CD20 antibody compositions
TW202321281A (en) Protease-mediated target specific cytokine delivery using fusion polypeptide

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: 23816908

Country of ref document: EP

Kind code of ref document: A1