US20160058884A1 - Methods for formulating antibody drug conjugate compositions - Google Patents

Methods for formulating antibody drug conjugate compositions Download PDF

Info

Publication number
US20160058884A1
US20160058884A1 US14/843,769 US201514843769A US2016058884A1 US 20160058884 A1 US20160058884 A1 US 20160058884A1 US 201514843769 A US201514843769 A US 201514843769A US 2016058884 A1 US2016058884 A1 US 2016058884A1
Authority
US
United States
Prior art keywords
antibody
drug
concentration
target
dar
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/843,769
Other languages
English (en)
Inventor
Gillian Payne
Robert W. Herbst
Juma Bridgewater
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunogen Inc
Original Assignee
Immunogen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunogen Inc filed Critical Immunogen Inc
Priority to US14/843,769 priority Critical patent/US20160058884A1/en
Assigned to IMMUNOGEN, INC. reassignment IMMUNOGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRIDGEWATER, JUMA, HERBST, ROBERT W., PAYNE, GILLIAN
Publication of US20160058884A1 publication Critical patent/US20160058884A1/en
Priority to US15/678,505 priority patent/US10603388B2/en
Priority to US16/407,384 priority patent/US20190365917A1/en
Priority to US18/146,307 priority patent/US20230364252A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68033Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a maytansine
    • A61K47/48569
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K47/48384
    • A61K47/48715
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68031Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68035Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a pyrrolobenzodiazepine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • 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
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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
    • C07K2317/90Immunoglobulins 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • Antibodies that specifically bind tumor surface antigens are used to deliver cytotoxic drugs in the form of antibody drug conjugates (ADCs).
  • Cytotoxic drugs are typically conjugated to antibodies at cysteine or lysine residues.
  • the number of molecules of a drug conjugated per antibody also termed the drug-to-antibody ratio (“DAR”), is typically a distribution of species ranging from 0-8.
  • DAR drug-to-antibody ratio
  • the DAR for a manufacturing batch of ADC is determined empirically using spectrophotometric measurements and ADC therapeutic compositions typically contain a mixture of ADC species that differ in drug load.
  • the DAR for an ADC hatch represents the average DAR of the ADC species within the batch.
  • ADC cancer therapeutics and antibody cancer therapeutics are both formulated based on nominal antibody protein concentration and must conform to specification. While the drug product label gives information about the “nominal” or target protein concentration, the drug concentration in the vial may vary relative to the target antibody concentration because of the allowed variation in DAR, even while conforming to the acceptance criteria.
  • the potency of ADCs is generally linear relative to concentration. Unlike antibodies, ADCs have an additional potential for variable potency due to the DAR.
  • a typical specification for antibody concentration and DAR allows the concentration of cytotoxic drug in the ADC product vial to vary somewhat from batch-to-batch when patient dosing is based on nominal antibody concentration.
  • ADC compositions would advantageously reduce variability in potency, efficacy, and/or toxicity between batches and ensure that patients receive an ADC within the intended therapeutic range.
  • the current invention provides a novel method for formulating a therapeutic composition comprising an antibody drug conjugate (“ADC”) based on drug concentration, thereby narrowing variability in potency between batches of ADC, minimizing toxicity and increasing the efficacy of drug formulated according to this method.
  • ADC antibody drug conjugate
  • the invention is based, at least in part, on the discovery that the efficacy and toxicity of some ADCs is driven entirely or predominantly by the concentration of drug administered rather than by antibody concentration.
  • Conventional methods of formulating an antibody based therapeutic, including ADC containing pharmaceutical compositions have relied on dosing the patient based on antibody concentration. While this might be advantageous to compositions that only contain an antibody, formulating ADCs using antibody concentrations can cause the drug concentrations to vary and potentially fall outside the desired range.
  • the DAR, antibody and drug each vary within a given and acceptable range, as defined by a given ADC specification.
  • the variability of one component affects the other.
  • a variability of ⁇ 10-20% in the antibody concentration which is within the acceptable range by industry standards, would cause a ⁇ 10-20% variation in the drug concentration which can cause a ⁇ 20-40% variability of potency of the drug product in the vial.
  • a ⁇ 15% variation in DAR would add further variation in potency as it would allow 15% higher or lower concentrations of drug. This effect can be particularly relevant for a specific subset of ADCs where it has been demonstrated that the concentration of the drug is the main driver of the toxicity and efficacy.
  • ADCs are linked to cytotoxic agents, also known as “drug” molecules and the number of drug molecules conjugated per antibody molecule is represented by the term ‘drug-to-antibody’ ratio (“DAR”).
  • DAR drug-to-antibody ratio
  • the DAR for a manufacturing batch of ADC is determined empirically using spectrophotometric measurements by obtaining the ratio of concentration of drug to that of antibody.
  • the DAR for a particular batch of ADC represents an average number of drugs attached to each antibody molecule within that batch. Typical DAR specifications for clinical development are in the range of ⁇ 10-15%.
  • the initial step in an ADC formulation is to determine the molar concentration for both the drug and the antibody and to calculate the DAR.
  • the ADC is then formulated to a target antibody concentration allowing the drug concentration to vary according to the manufactured DAR value, as follows:
  • the present invention is based on the discovery that, by formulating the ADC composition based on a target drug concentration defined at a fixed antibody concentration and fixed DAR, the variability in potency and toxicity can be minimized. Therefore, in cases where it has been demonstrated that the potency, efficacy and/or toxicity of the ADC are primarily driven by the amount of drug administered, improved methods for reducing variability of the cytotoxic drug concentration would be beneficial. Accordingly, the formulation methods described below include determining target drug concentration at a feed antibody concentration and a fixed DAR and formulating the antibody drug conjugate composition to achieve the target drug concentration. Such improved formulation methods ensure that patients are dosed within a narrow intended drug range without additional risk of batch failure.
  • the invention generally provides a method of reducing (e.g., by at least about 5%, 10%, 20% or more) potency variability in an antibody drug conjugate composition, the method involving determining target drug concentration at a fixed antibody concentration and drug antibody ratio; and formulating the antibody drug conjugate composition to achieve the target drug concentration, thereby reducing potency variability in the composition.
  • the variability in the drug concentration is about ⁇ 10%.
  • the variability is less than about ⁇ 5, 6, 7, 8, or 9%.
  • the method reduces batch-to-batch potency variation (e.g., by at least about 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, or more).
  • the composition is a finished drug product.
  • the drug concentration varies within the antibody specification concentration.
  • the antibody concentration is equal to target antibody concentration ⁇ less than about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%.
  • antibody concentration is equal to target antibody concentration ⁇ less than, about 10%, 12%, 15%, or 20%.
  • the invention provides a method of reducing potency variability in a composition comprising an antibody drug conjugate.
  • the method involves formulating the antibody drug conjugate by targeting a variable drug concentration which falls in the midpoint of the range where the antibody concentration specification range and the drug concentration specification range overlap, thereby reducing potency variability in the composition.
  • the invention provides a method of reducing potency variability in a composition containing an antibody drug conjugate involving: (a) measuring the DAR for the antibody drug conjugate composition; (b) determining the upper antibody specification limit and the lower antibody specification limit, where the upper antibody specification limit is the target antibody concentration plus the maximum variation allowed by the specification and the lower antibody specification limit is the target antibody concentration minus the maximum variation allowed by the specification; (c) determining the defined upper drug specification limit and the defined lower drug specification limit, where the defined upper drug specification limit is the target drug concentration plus the maximum variation allowed by the specification and the defined lower drug specification limit is the target drug concentration minus the maximum variation allowed by the specification; (d) determining the calculated upper drug specification limit (USL (drug)) as follows:
  • step (f) comparing the calculated USL (drug) of step (d) to the defined upper drug specification limit of step (c), and selecting the lower of the two values as the effective upper drug specification limit;
  • step (g) comparing the calculated LSL (drug) of step (e) to the defined lower drug specification limit of step (c), and selecting the higher of the two values as the effective lower drug specification limit; and
  • step (h) formulating the antibody drug conjugate composition to a target drug concentration that is the midpoint between the effective upper drug specification limit and the effective lower drug specification limit, thereby reducing potency variability in the composition.
  • the method narrows the range of the upper and lower specification limits for the drug to about ⁇ 3-9%.
  • the method narrows the range of the upper and lower specification limits for the drug to about ⁇ 4%.
  • the maximum variation allowed by the specification in step (b) is about ⁇ 15%, In another embodiment, the maximum variation allowed by the specification in step (b) is less than about ⁇ 10, 11, 12, 13, or 14%. In one embodiment, the maximum variation allowed by the specification in step (c) is about ⁇ 15%. In another embodiment, the maximum variation allowed by the specification in step (c) is less than about ⁇ 10, 11, 12, 13, or 14%.
  • the antibody is a non-functional antibody.
  • the DAR is at the lower limit of DAR specification or at the upper limit of DAR specification. In various embodiments, the lower limit of DAR specification is 2.3, 2.4, or 2.5. In various embodiments, the upper limit of DAR specification is 2.9, 3.0, or 3.1.
  • the invention also provides a method of formulating an antibody drug conjugate by targeting a variable antibody concentration which falls in the midpoint of the range where the antibody concentration specification range and the drug concentration specification range overlap, thereby targeting an antibody concentration that will allow the least fluctuation in the drug concentration when the ADC is formulated. This allows for tighter control of the antibody concentration in the ADC formulation.
  • the invention provides a method of formulating an antibody drug conjugate composition, the method involving determining target drug concentration at a fixed antibody concentration and drug antibody ratio; and formulating the antibody drug conjugate composition to achieve the target drug concentration.
  • the invention provides a method of reducing potency variability in a composition comprising an antibody maytansinoid conjugate, the method involving determining target maytansinoid concentration at a fixed antibody concentration and maytansinoid-to-antibody ratio; and formulating the antibody maytansinoid conjugate composition to achieve the target maytansinoid concentration, thereby reducing potency variability in the composition.
  • the invention provides a method of formulating an antibody maytansinoid conjugate composition, the method involving determining target maytansinoid concentration at a fixed antibody concentration and maytansinoid-to-antibody ratio; and formulating the antibody maytansinoid conjugate composition to achieve the target maytansinoid concentration.
  • the invention provides a method of formulating an antibody benzodiazepine (e.g., pyrrolobenzodiazepine or indolinobenzodiazepine) conjugate composition, the method involving determining target benzodiazepine concentration at a fixed antibody concentration and benzodiazepine-to-antibody ratio; and formulating the antibody benzodiazepine conjugate composition to achieve the target benzodiazepine concentration.
  • an antibody benzodiazepine e.g., pyrrolobenzodiazepine or indolinobenzodiazepine
  • the invention provides a method of reducing potency variability in a composition containing an antibody benzodiazepine (e.g., pyrrolobenzodiazepine or indolinobenzodiazepine) conjugate involving; (a) measuring the DAR for the antibody benzodiazepine (e.g., pyrrolobenzodiazepine or indolinobenzodiazepine) conjugate composition; (b) determining the upper antibody specification limit and the lower antibody specification limits where the upper antibody specification limit is the target antibody concentration plus the maximum variation allowed by the specification and the lower antibody specification limit is the target antibody concentration minus the maximum variation allowed by the specification; (c) determining the defined upper benzodiazepine specification limit and the defined lower benzodiazepine specification limit, where the defined upper benzodiazepine specification limit is the target benzodiazepine concentration plus the maximum variation allowed by the specification and the defined lower benzodiazepine specification limit is the target benzodiazepine concentration minus the maximum variation allowed by the specification;
  • step (f) comparing the calculated USL (drug) of step (d) to the defined upper benzodiazepine specification limit of step (c), and selecting the lower of the two values as the effective upper benzodiazepine specification limit;
  • step (g) comparing the calculated LSL (drug) of step (e) to the defined lower benzodiazepine specification limit of step (c), and selecting the higher of the two values as the effective lower benzodiazepine specification limit; and
  • step (h) formulating the antibody benzodiazepine conjugate composition to a target benzodiazepine concentration that is the midpoint between the effective upper benzodiazepine specification limit and the effective lower benzodiazepine specification limit, thereby reducing potency variability in the composition.
  • the invention provides a method for dosing a subject within a narrow intended range, the method involves providing an antibody drug conjugate composition formulated according to the method of any previous aspect, and administering said composition to the subject.
  • the invention provides a pharmaceutical composition containing an antibody drug conjugate formulated according to the method of a previous aspect, where a nominal drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration is provided on the label.
  • a nominal drug e.g., maytansinoid, benzodiazepine compounds, auristatin
  • the drug is a cytotoxic agent.
  • Cytotoxic agents include, without limitation, tubulin inhibitors, DNA damaging agents, DNA cross linkers, DNA alkylating agents, and cell cycle or mitotic disrupters.
  • Non-limiting examples of cytotoxic agents include maytansinoids; benzodiazepine compounds, such as pyrrolobenzodiazepines and indolinobenzodiazepines; and auristatins).
  • the method reduces batch-to-batch potency variation (e.g., by at least about 5%, 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50% or more).
  • the composition is a finished drug product.
  • the antibody concentration varies within the antibody specification concentration.
  • the method reduces batch-to-batch potency variability in producing the antibody maytansinoid conjugate.
  • the composition is allowed to vary in potency by about 10-40% (e.g., 10, 15, 20, 25, 30, 35, 40%).
  • the composition is allowed to vary in potency by about 10-20% (e.g., 10, 12, 15, 18, 20%).
  • the antibody concentration specification is equal to target ⁇ less than shout 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%.
  • antibody concentration specification is equal to target ⁇ less than about 10%, 12%, 15%, or 20%.
  • variability in composition potency is reduced relative to when the antibody drug conjugate composition is formulated based on antibody concentration.
  • the antibody concentration and conjugated drug e.g., maytansinoid, benzodiazepine compounds, auristatin
  • concentration are determined by spectrophotometric measurement.
  • drug to antibody ratio is determined by size exclusion chromatography (SEC) or by SEC-mass spectrometry (SEC-MS).
  • the efficacy or toxicity of the composition is independent of drug to antibody ratio or antibody concentration, but dependent on the total administered dose of conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin).
  • conjugated drug e.g., maytansinoid, benzodiazepine compounds, auristatin
  • efficacy of the composition is independent of or largely independent of drug to antibody ratio.
  • toxicity of the composition is independent of or largely independent of drug to antibody ratio.
  • efficacy or toxicity depends on or largely depends on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration.
  • efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration.
  • toxicity depends, substantially depends, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration.
  • efficacy is independent of, substantially independent of, or is at least in part independent of antibody concentration.
  • toxicity is independent of substantially independent of, or is as least in part independent of antibody concentration.
  • efficacy and toxicity depend on, substantially depend on, or depend at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration. In various embodiments of the previous aspects, efficacy and toxicity depend less on antibody concentration than on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration. In various embodiments of the previous aspects, efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration.
  • conjugated drug e.g., maytansinoid, benzodiazepine compounds, auristatin
  • toxicity depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration.
  • efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration and toxicity depends on, substantially depends on, or depends at least in part on conjugated drug concentration.
  • efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration and toxicity depends on, substantially depends on, or depends at least in part on antibody concentration.
  • toxicity depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration and on antibody concentration
  • efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration.
  • toxicity depends on, substantially depends on, or depends at least in part on antibody concentration and efficacy depends on, substantially depends on, or depends at least in part on conjugated drug (e.g., maytansinoid, benzodiazepine compounds, auristatin) concentration.
  • conjugated drug e.g., maytansinoid, benzodiazepine compounds, auristatin
  • the antibody drug conjugate composition is formulated for infusion.
  • the antibody drug conjugate is formulated with a pharmaceutically acceptable parenteral vehicle.
  • the antibody drug conjugate is formulated in a unit dosage injectable form.
  • the method comprises determining an upper specification limit (USL) and a lower specification limit (LSL).
  • USL upper specification limit
  • LSL lower specification limit
  • the cytotoxic compound or drug is a tubulin inhibitor, DNA damaging agent, DNA cross linker, DNA alkylating agent, or cell cycle or mitotic disrupter.
  • a drug includes, but is not limited to, maytansinoids and maytansinoid analogs, benzodiazepine compounds (e.g., pyrrolobenzodiazepines and indolinobenzodiazepines; see also Table 1: compounds D1-D10 and DGN462), taxoids, CC-1065 and CC-1065 analogs, duocarmycins and duocarmycin analogs, enediynes, such as calicheamicins, dolastatin and dolastatin analogs including auristatins, tomaymycin derivatives, leptomycin derivatives, methotrexate, cisplatin, carboplatin, daunorubicin, doxorubicin, vincristine, vinblastine, melphalan, mitomycin C, chlorambucil, and morpholino-doxorubicin.
  • benzodiazepine compounds e.g., pyrrolobenzodiazepines and indolinobenzodiazepines;
  • the maytansinoid is DM1, DM3, or DM4.
  • the benzodiazepine compounds are selected from the representative cytotoxic agents D1-D10 and DGN462 listed in Table 1 below.
  • the antibody may be a functional antibody or non-functional antibody.
  • Non-functional antibodies include, for example, huDS6 and antibodies with only effector-mediated cell killing, such as huMov19 (M9346A), huAnti-CD123, huMy9-6 (Z4681A), and huB4.
  • Functional antibodies include, for example, huEGFR-7R and huCD37-3.
  • the drug is a benzodiazepine compound and the antibody is a non-functional antibody.
  • the drug is a maytansinoid and the antibody is a non-functional antibody.
  • the linker is a cleavable linker, such as N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP), N-succinimidyl 4-(2-pyridyldithio)butanoate (SPDB), N-succinimidyl 4-(2-pyridyldithio)2-sulfobutanoate (sulfo-SPDB), or N-succinimidyl 4-(2-pyridyldithio)pentanoate (SPP).
  • SPDP N-succinimidyl 3-(2-pyridyldithio) propionate
  • SPDB N-succinimidyl 4-(2-pyridyldithio)butanoate
  • SPP N-succinimidyl 4-(2-pyridyldithio)pentanoate
  • the linker is a non-cleavable linker, such as 2-iminothiolane, acetylsuccinic anhydride, succinimidyl 4-(maleimidomethyl)cyclohexanecarboxylate (SMCC).
  • SMCC succinimidyl 4-(maleimidomethyl)cyclohexanecarboxylate
  • the generic linkers 2-iminothiolane and acetylsuccinic anhydride can be used as cleavable or non-cleavable linkers.
  • the linker antibody drug conjugate is huMov19-sulfo-SPDB-DM4, huMov19-sulfo-SPDB-D1, huMov19-D2, huMov19-sulfo-SPDB-D10, huMov19-sulfo-SPDB-DGN462, huMy9-6-sulfo-SPDB-D1, huMy9-6-D2, huMy9-6-sulfo-SPDB-D10, huMy9-6-sulfo-SPDB-DGN462, huAnti-DC123-sulfo-SPDB-D1, huAnti-DC123-D2, huAnti-CD123-sulfo-SPDB-D10, huAnti-CD123-sulfo-SPDB-DGN462, huB4-SPDB-DM4, huDS6-SPDB-DM4,
  • AIBW adjusted ideal body weight
  • IBW, LBW, and ADJ are discussed in more detail in Green and Duffull, British Journal of Clinical Pharmacology 58: 119-133 (2004), which is herein incorporated by reference in its entirety.
  • cytotoxic agent is meant a small molecule chemical compound, peptide, or nucleic acid molecule that is toxic to cells.
  • drug is used to refer to a cytotoxic agent.
  • the term “drug” is used interchangeably with the term “cytotoxic agent.”
  • the cytotoxic agent is conjugated to an antibody.
  • the cytotoxic agent is a maytansinoid, such as DM1, DM3, or DM4.
  • cytotoxic agents include, but are not limited to, benzodiazepine compounds (e.g., pyrrolobenzodiazepines and indolinobenzodiazepines; see also Table 1; compounds D1-D10 and DGN462), taxoids, CC-1065 and CC1065 analogs, duocarmycins and duocurmycin analogs, enediynes, such as calicheamicins, dolastatin and dolastatin analogs including auristatins, tomaymycin derivatives, leptomycin derivatives, methotrexate, cisplatin, carboplatin, daunorubicin, doxorubicin, vincristine, vinblastine, melphalan, mitomycin C, chlorambucil, and morpholino-doxorubicin.
  • benzodiazepine compounds e.g., pyrrolobenzodiazepines and indolinobenzodiazepines; see also Table 1; compounds D1-D10 and DGN
  • DAR drug-to-antibody ratio
  • MAR maytansinoid-to-antibody ratio
  • target antibody concentration is meant a desired antibody concentration
  • target drug concentration or “target cytotoxic agent concentration” is meant a desired concentration of a drug or cytotoxic agent. It should be noted that the concentration of drug or cytotoxic agent is predominantly calculated based on the conjugated form of the drug but may include minor amounts of free or unconjugated drug found in the sample.
  • target maytansinoid concentration is meant a desired concentration of maytansinoid.
  • Potency variability is meant the different potencies present in different batches of drug product. Potency variability is desirably reduced by at least about 5%, 10%, 20%, 25%, 30%, 40%, 50% or more.
  • a finished drug product is a container (e.g., vial) that contains an antibody drug conjugate of the invention, alone or in combination with an excipient.
  • Specification is meant a set of criteria to which a drug or drug product must conform to be acceptable for its intended use.
  • a specification is typically proposed by a manufacturer and agreed to by a regulatory body (e.g., the FDA).
  • “functional antibody” is meant to refer to an antibody that affects cell death by a direct cell killing mechanism, such as apoptosis or necrosis. Functional antibodies have direct cell killing activity in vivo without being conjugated to a drug (“naked antibody”). Non-limiting examples of functional antibodies include the huEGFR-7R antibody and the huCD37-3 antibody.
  • non-functional antibody is meant to refer to an antibody that has (i) no known cell killing activity in vivo (e.g., no direct or indirect cell killing as a naked antibody, for example, huDS6) or (ii) indirect cell killing activity as a result of effector function, for example, antibody-dependent cell-mediated cytotoxicity (ADCC), antibody dependent cellular phagocytosis (ADCP) and complement dependent cytotoxicity (CDC), or (iii) has increased conjugate activity in vivo when effector function is increased or any combination of (i), (ii), and (iii).
  • a non-functional antibody may have anti-proliferative activity, for example, by blocking binding of a proliferative agent (e.g., growth factor).
  • Non-limiting examples of non-functional antibodies that have indirect cell killing activity include huMov19, huMy9-6, and huB4.
  • huB4 is meant a humanized antibody or epitope binding fragment thereof that specifically binds CD19, such as human CD9.
  • An exemplary huB4 antibody of the invention may include the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huB4-SPDB-DM4 an antibody drug conjugate that includes an huB4 antibody, which specifically binds CD19, conjugated to the cytotoxic maytansinoid, N 2′ -deacetyl-N 2′ -(4-mercapto-4methyl-1-oxopentyl) maytansine (DM4) via the linker N-succinimidyl 4-(2-pyridyldithio)butanoate (SPDB).
  • huB4-SPDB-DM4 is described, for example, in U.S. Pat. No. 8,435,528 and International Pat. Appl. Publication No. WO2004/103272, which are incorporated herein by reference in their entireties.
  • huMov19 (also termed “M9346A”) is meant a humanized antibody or epitope binding fragment thereof that specifically binds folate receptor alpha (also known as folate receptor 1 or “FOLR1” herein).
  • folate receptor alpha also known as folate receptor 1 or “FOLR1” herein.
  • Detailed sequences for huMov19 are described in U.S. Pat. Nos. 8,557,966 and 8,709,432 and International Pat. Appl. Publication Nos.: WO2011/106528, which are incorporated herein by reference in their entireties.
  • Exemplary huMOV19 antibodies of the invention may include the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huMov19-sulfo-SPDB-DM4 also termed “IMGN853” is meant an antibody drug conjugate that comprises an huMov19 antibody, which specifically binds FOLR1, conjugated to the cytotoxic maytansinoid, N 2′ -deacetyl-N 2′ -(4-mercapto-4-methyl-1-oxopentyl) maytansine (DM4) via the disulfide-containing linker N-succinimidyl 4-(2-pyridyldithio)-2-sulfobutanoate (sSPDB).
  • DM4 conjugated to the cytotoxic maytansinoid
  • sSPDB disulfide-containing linker N-succinimidyl 4-(2-pyridyldithio)-2-sulf
  • the ADC huMov19-sulfo-SPDB-DM4 is described, for example, by Ab et al., AACR; Cancer Res 2011;71(8 Suppl):Abstract number 4576, and U.S. Pat. Nos. 8,557,966 and 8,709,432 and International Pat. Appl. Publication Nos.: WO2011/106528 which are each incorporated herein by reference in their entirety.
  • huDS6 is meant a humanized antibody or epitope binding fragment thereof that specifically binds a CA6 siaioglycotope on the Mucl mucin receptor (e.g., human Mucl) expressed by cancerous cells.
  • exemplary sequences for huDS6 are described in U.S. Pat. No. 7,834,155 and International Pat. Appl. Publication Nos.: WO2005/009369 and WO2007/024222, which are incorporated herein by reference in their entireties.
  • An exemplary huDS6 antibody of the invention may include or consists of the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huMy9-6 (also termed “Z4681A”) is meant a humanized antibody or epitope binding fragment thereof that specifically binds leukocyte differentiation antigen CD33, such as human CD33.
  • exemplary sequences for the huMy9-6 heavy chain variable region portion are described in U.S. Patent Publication No. 20060177455, which is incorporated herein by reference in its entirety.
  • Exemplary sequences for the huMy9-6 light chain variable region portion are known in the art and described in U.S. Pat. Nos. 7,557,189, 7,342,110, 8,119,787 and 8,337,855, which are incorporated herein by reference in their entireties.
  • An exemplary huMy9-6 antibody of the invention may include or consists of the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huMy9-6-sulfo-SPDB-DGN462 (also termed “IMGN779”) is meant an anti-huCD33 antibody conjugated to an indolinobenzodiazepine dimer containing a mono-imine moiety termed DGN462 via cleavable disulfide linker.
  • huEGFR-7R also termed “J2898A” is meant a humanized antibody or epitope binding fragment thereof that specifically binds EGFR, such as human EGFR.
  • An exemplary huEGFR-7R antibody of the invention may include or consists of the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huEGFR-7R-SMCC-DM1 an antibody drug conjugate that comprises an huEGFR-7R antibody, which specifically binds EGFR, conjugated to the maytansinoid N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine (DM1) via the linker N-succinimidyl 4-(maleimidomethyl)cyclohexanecarboxylate (SMCC).
  • DM1 maytansinoid N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine
  • DM1 maytansinoid N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine
  • DM1 maytansinoid N(2′)-deacetyl-N(2′)-(3
  • huCD37-3 is meant a humanized antibody or epitope binding fragment thereof that specifically binds CD37, such as human CD37.
  • Exemplary sequences for huCD37-3 are described in U.S. Pat. No. 8,765,917 and International Pat. Appl. Publication. No. WO2011/112978, which are incorporated herein by reference in their entireties.
  • An exemplary huCD37-3 antibody of the invention may include or consists of the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huCD37-3-SMCC-DM1 (also termed “IMGN529”) is meant an antibody drug conjugate that comprises a humanized IgG1 antibody K7153A that specifically binds CD37 that is covalently linked via the uncleavable, maleimide-derived thioether-based linker succinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate (SMCC) to the maytansinoid N(2′)-deacetyl-N(2′)-(3-mercapto-1-oxopropyl)-maytansine (DM1).
  • SMCC N-maleimidomethyl]cyclohexane-1-carboxylate
  • huCD37-50 is meant a humanized antibody or epitope binding fragment thereof that specifically binds CD37, such as human CD37.
  • Exemplary sequences for huCD37-50 are described in U.S. Pat. No. 8,765,917 and International Pat. Appl. Publication No. WO2011/112978, which are incorporated herein by reference in their entireties.
  • An exemplary huCD37-50 antibody of the invention may include or consists of the following CDRs (shown in bold and underline) or the following light chain (LC) and heavy chain (HC) sequences:
  • huAnti-CD123 is meant a humanized antibody or epitope binding fragment thereof that specifically binds CD123, such as human CD123.
  • exemplary huAnti-CD123 antibodies are described in U.S. Provisional Appl. Ser. No, 62/186,161, which is incorporated herein by reference in its entirety.
  • antibody means an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • antibody encompasses intact polyclonal antibodies, intact monoclonal antibodies, epitope binding antibody fragments (such as Fab, Fab′, F(ab′)2, and Fv fragments), single chain Fv (scFv) mutants, immunoglobulin new antigen receptor antibodies (IgNARs), which comprise single variable new antigen receptor domain antibody fragments (V NAR S or Y NAR domains), unibodies, in which the hinge region has been removed, nanobodies, antibody fragments consisting of a single monomeric variable antibody domain (Ablynx), minibodies, which are engineered antibody fragments comprising an scFv linked to a CH domain (Hu et al., Cancer Res.
  • epitope binding antibody fragments such as Fab, Fab′, F(ab′)2, and Fv fragments
  • scFv single chain Fv mutants
  • IgNARs immunoglobulin new antigen receptor antibodies
  • V NAR S or Y NAR domains single variable new antigen receptor
  • DuoBodies® which are bispecific modified IgG1 antibodies that include (i) a stable hinge region that is non-permissive for Fab arm exchange in vivo and (ii) an IgG4-like CH3 domain modified to be permissive for Fab arm exchange in vivo.
  • multispecific antibodies such as bispecific antibodies generated from at least two intact antibodies, probodies, which are recombinant, masked monoclonal antibodies that remain inert in healthy tissue, but are activated specifically in the disease microenvironment (e.g., cleavage by a protease enriched or specific in a disease microenvironment)
  • disease microenvironment e.g., cleavage by a protease enriched or specific in a disease microenvironment
  • chimeric antibodies e.g., cleavage by a protease enriched or specific in a disease microenvironment
  • chimeric antibodies e.g., humanized antibodies, human antibodies, fusion proteins comprising an antigen determination portion of an antibody, and any other modified immunoglobulin molecule comprising an antigen recognition site so long as the antibodies exhibit the desired biological activity.
  • An antibody can be of any the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g. IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively.
  • the different classes of immunoglobulins have different and well known subunit structures and three-dimensional configurations.
  • a “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions.
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies.
  • antibody fragment refers to a portion of an intact antibody and refers to the antigenic determining variable regions of an intact antibody.
  • antibody fragments include, but are not limited to Fab, Fab′, F(ab′)2, and Fv fragments, linear antibodies, single chain antibodies, and multispecific antibodies formed from antibody fragments.
  • cancer refers to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth.
  • Cancer can include a hematological cancer or a solid tumor. More specifically, the cancer is leukemia (e.g., acute myeloid leukemia (AML), acute monocytic leukemia, promyelocytic leukemia, eosinophilic leukaemia, acute lymphoblastic leukemia (ALL) such as acute B lymphoblastic leukemia (B-ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL) or lymphoma (e.g., non-Hodgkin lymphoma), myelodysplastic syndrome (MDS), melanoma, lung cancer (e.g., non-small cell lung cancer; NSCLC), ovarian cancer, endometrial cancer, peritoneal cancer, pancreatic cancer, breast cancer, prostate cancer, squamous cell
  • AML acute mye
  • an analog is meant a molecule that is not identical but has analogous functional or structural features.
  • a polypeptide analog retains the biological activity of a corresponding naturally-occurring polypeptide, while having certain biochemical modifications that enhance the analog's function relative to a naturally occurring polypeptide. Such biochemical modifications could increase the analog's protease resistance, membrane permeability, or half-life, without altering, for example, ligand binding.
  • An analog may include an unnatural amino acid.
  • chimeric antibodies refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e.g. mouse, rat, rabbit, etc.) with the desired specificity, affinity, and capability while the constant regions are homologous to the sequences in antibodies derived from another (usually human) to avoid eliciting an immune response in that species.
  • Detect refers to identifying the presences absence or amount of the analyte to be defected.
  • disease is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • diseases include neoplasias and cancers to be treated with a composition of the invention.
  • an effective amount is meant the amount of an agent required to ameliorate the symptoms of a disease relative to an untreated patient.
  • the effective amount of active agent(s) e.g., an antibody drug conjugate (ADC) or drug
  • ADC antibody drug conjugate
  • drug used to practice the present invention for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an “effective” amount.
  • epitopes or “antigenic determinant” are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody.
  • the antigen is a polypeptide
  • epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • formulate is meant a process used to produce a drug product.
  • humanized antibody refers to forms of non-human (e.g. murine) antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human (e.g., murine) sequences.
  • humanized antibodies are human immunoglobulins in which residues from the complementary determining region (CDR) are replaced by residues from the CDR of a non-human species (e.g. mouse, rat, rabbit, hamster) that have the desired specificity, affinity, and capability (Jones et al., 1986, Nature, 321:522-525; Riechmann et al., 1988, Nature, 332:323-327; Verhoeyen et al., 1988.
  • CDR complementary determining region
  • the Fv framework region (FR) residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, affinity, and capability.
  • the humanized antibody can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or capability.
  • the humanized antibody will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the non-human immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. No. 5,225,539.
  • the goal of humanization is a reduction in the immunogenicity of a xenogenic antibody, such as a murine antibody, for introduction into a human, while maintaining the full antigen binding affinity and specificity of the antibody.
  • Humanized antibodies may be produced using several technologies, such as resurfacing and CDR grafting.
  • the resurfacing technology uses a combination of molecular modeling, statistical analysis and mutagenesis to alter the non-CDR surfaces of antibody variable regions to resemble the surfaces of known antibodies of the target host.
  • (1) position alignments of a pool of antibody heavy and light chain variable regions are generated to give a set of heavy and light chain variable region framework surface exposed positions wherein the alignment positions for all variable regions are at least about 98% identical; (2) a set of heavy and light chain variable region framework surface exposed amino acid residues is defined for a rodent antibody (or fragment thereof); (3) a set of heavy and light chain variable region framework surface exposed amino acid residues that is most closely identical to the set of rodent surface exposed amino acid residues is identified; (4) the set of heavy and light chain variable region framework surface exposed amino acid residues defined in step (2) is substituted with the set of heavy and light chain variable region framework surface exposed amino acid residues identified in step (3), except tor those amino acid residues that are within 5 angstroms of any atom of any residue of the complementarity-determining regions of the rodent antibody; and (5) the humanized rodent antibody having binding specificity is produced.
  • Antibodies can be humanized using a variety of other techniques including CDR-grafting (EP 0 239 400; WO 91/09967; U.S. Pat. Nos. 5,530,101; and 5,585,089), veneering or resurfacing (EP 0 592 106; EP 0 519 596; Padlan E. A., 1991, Molecular Immunology 28(4/5):489-498; Studnicka G. M. et al., 1994, Protein Engineering 7(6):805-814; Roguska M. A. et al., 1994, PNAS 91:969-973), and chain shuffling (U.S. Pat. No. 5,565,332).
  • Human antibodies can be made by a variety of methods known in the art including phage display methods. See also U.S. Pat. Nos. 4,444,887, 4,716,111, 5,545,806, and 5,814,313; and International Pat. Appl. Publication Nos.: WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741 (said references incorporated by reference in their entireties).
  • human antibody means an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art. This definition of a human antibody includes intact or full-length antibodies, fragments thereof, and/or antibodies comprising at least one human heavy and/or light chain polypeptide such as, for example, an antibody comprising murine light chain and human heavy chain polypeptides.
  • antibody drug conjugate refers to a compound that is linked to a cell binding agent (i.e., an antibody or fragment thereof).
  • a cell binding agent i.e., an antibody or fragment thereof.
  • the cell binding agent e.g., antibody
  • the linker is covalently bound to the drug by a linker.
  • isolated refers to material that is free to varying degrees from components which normally accompany it as found in its native state. “Isolate” denotes a degree of separation from original source or surroundings. “Purify” denotes a degree of separation that is higher than isolation.
  • a “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of this invention is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography.
  • the term “purified” can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel.
  • modifications for example, phosphorylation or glycosylation, different modifications may give rise to different isolated proteins, which can be separately purified.
  • a “linker” is any chemical moiety that is capable of linking a compound to a protein.
  • a linker links a drug, such as a maytansinoid, to a cell-binding agent, such as an antibody or a fragment thereof in a stable, covalent manner.
  • Linkers can be susceptible to or be substantially resistant to acid-induced cleavage, light-induced cleavage, peptidase-induced cleavage, esterase-induced cleavage, and disulfide bond cleavage, at conditions under which the compound or the antibody remains active.
  • Suitable linkers are well known in the art and include, for example, disulfide groups, thioether groups, acid labile groups, photolabile groups, peptidase labile groups and esterase labile groups. Linkers also include charged linkers, and hydrophilic forms thereof as described herein and known in the art.
  • Exemplary cleavable linkers include, but are not limited to: N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP), N-succinimidyl 4-(2-pyridyldithio)butanoate (SPDB), N-succinimidyl 4-(2-pyridyldithio)2-sulfobutanoate (sulfo-SPDB), and disulfide N-succinimidyl 4-(2-pyridyldithio)pentanoate (SPP).
  • SPDP N-succinimidyl 3-(2-pyridyldithio) propionate
  • SPDB N-succinimidyl 4-(2-pyridyldithio)butanoate
  • SPP disulfide N-succinimidyl 4-(2-pyridyldithio)pentanoate
  • non-cleavable linkers include, but are not limited to: 2-iminothiolane, acetylsuccinic anhydride, and succinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate (SMCC).
  • SMCC succinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate
  • the generic linkers 2-iminothiolane and acetylsuccinic anhydride can be used as cleavable or non-cleavable linkers.
  • a “monoclonal antibody” refers to a homogeneous antibody population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants.
  • the term “monoclonal antibody” encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (such as Fab, Fab′, F(ab′), Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
  • “monoclonal antibody” refers to such antibodies made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, and transgenic animals.
  • telomere binding By “specifically binds” is meant a compound or antibody that recognizes and binds a polypeptide of interest, but which does not substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the invention.
  • Nucleic acid molecules useful in the methods of the invention include any nucleic acid molecule that encodes a polypeptide of interest or a fragment thereof. Such nucleic acid molecules need not be 100% identical with an endogenous nucleic acid sequence, but will typically exhibit substantial identity. Polynucleotides having “substantial identity” to an endogenous sequence are typically capable of hybridizing with at least one strand of a double-stranded nucleic acid molecule. Nucleic acid molecules useful in the methods of the invention include any nucleic acid molecule that encodes a polypeptide of the invention or a fragment thereof. Such nucleic acid molecules need not be 100% identical with an endogenous nucleic acid sequence, but will typically exhibit substantial identity. Polynucleotides having “substantial identity” to an endogenous sequence are typically capable of hybridizing with at least one strand of a double-stranded nucleic acid molecule.
  • substantially identical is meant a polypeptide or nucleic acid molecule exhibiting at least 50% identify to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein).
  • a reference amino acid sequence for example, any one of the amino acid sequences described herein
  • nucleic acid sequence for example, any one of the nucleic acid sequences described herein.
  • such a sequence is at least 60%, more preferably 80% or 85%, and more preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
  • Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e ⁇ 3 and e ⁇ 100 indicating a closely related sequence.
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin
  • subject is meant a mammal, including, but not limited to, a human or non-human mammal such as a bovine, equine, canine, ovine, or feline.
  • therapeutically effective amount refers to an amount of an antibody or other drug effective to “treat” a disease or disorder in a subject or mammal.
  • the therapeutically effective amount of the drug can reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent or stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent or stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. See the definition herein of “treating”.
  • the drug can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • Ranges provided herein are understood to be shorthand for all of the values within the range.
  • a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50.
  • the terms “treat,” “treating,” “treatment,” and the like refer to reducing or ameliorating a disorder and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated.
  • the term “about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. “About” can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about.
  • variable in my definition of a variable herein includes definitions of that variable as any single group or combination of listed groups.
  • the recitation of an embodiment for a variable or aspect herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
  • compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
  • MAR maytansinoid-to-antibody ratio
  • FIG. 2 is a graph showing the dependence of cytotoxic potency on the concentration of huMov19-sulfo-SPDB-DM4.
  • FIG. 3 provides two scatter plots and a table that simulate the likely effect of drug-to-antibody ratio (DAR) on eye toxicity (“ocular tox”).
  • DAR drug-to-antibody ratio
  • AIBW refers to Adjusted Ideal Body Weight. The DARs were calculated based on drug dosing levels.
  • FIG. 4 provides a scatter plot and a table that simulates the likely effect of drug-to-antibody ratio (DAR) and concentration on eye toxicity.
  • DAR drug-to-antibody ratio
  • concentration on eye toxicity The DARs were calculated and the corresponding ADCs were not actually administered to patients.
  • FIG. 5 includes two graphs showing the lack of effect of DAR on median tumor volume in KB and IGROV-1 murine xenograft models when the huMov19-sulfo-SPDB-DM4conjugate is administered at the same DM4 dose.
  • Mice were dosed with huMov19-sSPDB-DM4. All conjugates were dosed at 25 ⁇ g/kg of DM4 and variable antibody dose (higher for the low DAR conjugates and lower for the high DAR conjugates). Similar anti-tumor activity was observed regardless of variable DAR and antibody dose.
  • FIG. 6 is a graph showing the similar effect of DM4 dose on murine body weight for a conjugate having 9.0 DAR compared with 3.6 DAR when the administered DM4 dose is the same. All conjugates were dosed at 1.4 mg/kg of DM4 and variable antibody dose (higher for the low DAR conjugates and lower for the high DAR conjugates).
  • FIG. 7 is a graph showing the similar effect of DM1 dose on mean body weight change for conjugates having various maytansinoid-to-antibody ratios when the administered DM1 dose is the same. All conjugates were dosed at 3.0 mg/kg of DM1 and variable antibody dose (higher for the low DAR conjugates and lower for the high DAR conjugates). Toxicity was similar for all conjugates regardless of DAR.
  • FIG. 8 is a graph showing in vivo toxicity studies in mice that received antibody-SPDB-DM4 conjugates of various DARs, including ADC huDS6-SPDB-DM4 (“huDS6-DM4”); huB4-SPDB-DM4 (“huB4-DM4”); and huMy9-6-SPDB-DM4 (“huMy9-6-DM4”).
  • FIGS. 9A and 9B are tables showing the advantage of preparing an ADC composition based on DM4 concentration.
  • FIG. 9A shows the allowable DM4 concentrations in ⁇ g/ml that results when an antibody drug conjugate is formulated at a target antibody concentration (5.0 ⁇ 1.0 mg/ml) and DAR 3.4 ⁇ 0.5 (circled).
  • the DMA concentration is 91.1 at the 5.0 mg/mL target antibody concentration and the 3.4 DAR target (box).
  • Lower potency variation boxed area of DM4 concentrations allowed by formulating based on DMA concentration with a ⁇ 10% specification.
  • Target DAR, antibody and DM4 concentrations are boxed.
  • FIG. 9B antibody concentration specification fails at the high-low DAR extremes (boxed areas).
  • Target DAR, antibody and DM4 concentrations are boxed.
  • FIGS. 10A-10C are graphs showing the effect of formulating an antibody drug conjugate (ADC) batch by varying antibody concentration to achieve a target drug (DGN462) concentration.
  • ADC antibody drug conjugate
  • DGN462 is an exemplary drug.
  • the vertical line (elongated “I”) denotes the upper and lower limits of drug concentration.
  • DAR denotes drug-to-antibody ratio.
  • FIGS. 11A-11C are graphs showing that formulating an ADC composition by varying antibody and drug concentration (open ovals) narrows the permitted specification range for both the antibody and the drug relative to varying only antibody specification to achieve a target drug concentration (gray diamonds).
  • the invention provides improved methods for formulating a therapeutic composition comprising an antibody drug conjugate (“ADC”), thereby narrowing variability in potency between batches of ADC and/or narrowing drug and antibody specifications over a broader drug-to antibody ratio (DAR) range.
  • ADC antibody drug conjugate
  • the invention is based, at least in part, on the discovery that the efficacy and toxicity of some ADCs is driven entirely or in part by the dose of drug administered rather than the dose of antibody.
  • Formulating ADC compositions based on a target drug concentration advantageously minimizes potency variations in the finished drug product and ensures that patients are dosed within a narrow intended range.
  • antibody drug conjugate therapeutic compositions have been formulated based on antibody concentration. Some variability is inherent in formulating antibody drug conjugates based on antibody concentration, even when remaining within the allowable ranges of a given specification.
  • concentration of antibody in the conjugate is measured, and the conjugate is diluted to reach the target drug concentration based on the fixed antibody concentration.
  • the antibody concentration in the finished drug product is allowed to vary from the target concentration.
  • the formulation specification allows ⁇ 20% variation in antibody-based concentration (e.g., 4.0-6.0 mg/mL allowed for a target antibody concentration, of 5.0 mg/mL).
  • the ADC potency in the finished drug product could vary by as much as ⁇ 35% and potentially fall outside the desired range.
  • the formulation methods reported herein below involve determining drug concentration at a fixed antibody concentration and fixed drug-to-antibody ratio and formulating the antibody drug conjugate composition to achieve a desired drug concentration.
  • formulating the ADC composition based on drug concentration and adding a drug concentration specification of, for example ⁇ 10% significantly narrows the potency present in the finished drug product to ⁇ 10% of the target drug concentration ( ⁇ 10% specification).
  • a drug concentration specification for example ⁇ 10% significantly narrows the potency present in the finished drug product to ⁇ 10% of the target drug concentration ( ⁇ 10% specification).
  • This formulation strategy only slightly increases the risk that antibody concentration will be outside specification, and thus the risk of batches failing to conform to specification is fairly low.
  • Such improved formulation methods ensure that patients are dosed within a narrow intended range without adding substantially to the risk that a batch of ADC will fail to conform to specification.
  • the invention provides a method of reducing potency variability in a composition comprising an antibody drug conjugate.
  • the method involves formulating the antibody drug conjugate by targeting a variable concentration of both the drug and the antibody (i.e. by having small variations ( ⁇ 4-9%) in both concentration values rather than large changes in one concentration ( ⁇ 10-15%) within a range where both specifications overlap, thereby reducing potency variability in the composition.
  • a small variation is about 4, 5, 6, 7, 8, or 9%.
  • a large change is about 10, 11, 12, 13, 14, or 15%.
  • the invention provides a method of reducing potency variability in a composition comprising an antibody drug conjugate.
  • the method involves formulating the antibody drug conjugate by targeting a variable concentration of either the drug or the antibody within a range where both specifications overlap, thereby reducing potency variability in the composition.
  • ADC cancer therapeutics are formulated similarly to antibody cancer therapeutics; that is, based on the antibody protein concentration. While the drug product label gives information about the “nominal” or target concentration, which is the basis for dosing (for instance on a mg/kg or mg/m 2 basis), a typical specification for antibody concentration is target ⁇ 10-20%. Potency of ADCs is generally linear relative to concentration, therefore the potency of a drug product may vary by ⁇ 20%.
  • DAR Drug to Antibody Ratio
  • the toxicity is entirely dependent on the dose of conjugated drug administered regardless of the dose of antibody.
  • the toxicity is independent of DAR as long as the administered dose of me conjugated drug is the same.
  • efficacy depends entirely on the dose of drug. In such cases, the efficacy is the same regardless of DAR as long as the administered dose of the conjugated drug is the same.
  • a typical specification for antibody concentration and DAR allows the concentration of the conjugated drug to vary somewhat.
  • the ADCs efficacy may still be driven more by the dose of drug rather than by the antibody.
  • the invention provides methods for formulating a therapeutic composition based on the concentration of the drug rather than the concentration of the antibody.
  • the target concentration for the drug would be the calculated drug concentration at a fixed antibody concentration and fixed DAR.
  • a specification set close to the target conjugated drug concentration would dictate the allowable potency variation in the drug product vial. Therefore, a specification for drug concentration of ⁇ 10% would narrow the allowable potency variation to ⁇ 10%.
  • a therapeutic composition may be formulated by targeting a variable drug concentration based on the DAR and antibody specification to achieve an ADC therapeutic composition that falls within the center of the effective specification range for antibody and the drug concentrations, where both the drug and antibody concentrations overlap.
  • DARs of within ⁇ 5% of target the center of effective range achieved by targeting a variable drug concentration is used. Improvements are realized when DAR varies between ⁇ 5-15% of target DAR.
  • formulating a therapeutic composition by targeting a variable drug concentration can provide smaller variability with respect to both drug (e.g., about ⁇ 4%) and antibody concentration (e.g., about ⁇ 10% ) versus targeting a static drug concentration, which would vary the antibody concentration by about ⁇ 15%.
  • Such ranges are useful in formulating ADC compositions, examples of which includes huMy9-6-sulfo-SPDB-DGN462.
  • the invention features the use of the methods described herein for formulating huMy9-6-sulfo-SPDB-DGN462, which is an antibody drug conjugate comprising DGN462 conjugated to the anti-CD33 antibody, huMy9-6, via a cleavable disulfide linker, s-SPDB.
  • Other drugs useful in the invention include benzodiazepines such as those represented in Table 1 or variations thereof, and by the following structural formulas:
  • the present invention is directed to improved methods for formulating an ADC, comprising an antibody (e.g., antibody that binds a tumor antigen) or antibody fragment, and their functional equivalents as disclosed herein, linked or conjugated to a cytotoxic agent (e.g., drug or prodrug).
  • an antibody e.g., antibody that binds a tumor antigen
  • antibody fragment e.g., antibody fragment, and their functional equivalents as disclosed herein, linked or conjugated to a cytotoxic agent (e.g., drug or prodrug).
  • a cytotoxic agent e.g., drug or prodrug.
  • the invention includes, but is not limited to, an ADC that includes any of the following antibodies: huMov19, huMy9-6, huAnti-CD123, huB4, huDS6, huCD37-50, huCD37-3, and huEGFR-7R.
  • Suitable drugs or prodrugs are known in the art.
  • the drugs or prodrugs can be cytotoxic.
  • a cytotoxic drug used in the ADCs of the present invention may be any compound that results in the death of a cell (e.g., cancer cell), or induces cell death, or in some manner decreases cell viability, and includes, for example, tubulin inhibitors, DNA damaging agents, DNA cross linkers, DNA alkylating agents, and cell cycle disrupters.
  • suitable cytotoxic drugs include maytansinoids and maytansinoid analogs.
  • cytotoxic drugs include, for example, benzodiazepines (e.g., pyrrolobenzodiazepines and indolinobenzodiazepines; see also Table 1: compounds D1-D10 and DGN462), taxoids, CC-1065 and CC1065 analogs, duocarmycins and duocarmycin analogs, enediynes, such as calicheamicins, dolastatin and dolastatin analogs including auristatins, tomaymycin derivatives, leptomycin derivatives, methotrexate, cisplatin, carboplatin, daunorubicin, doxorubicin, vincristine, vinblastine, melphalan, mitomycin C, chlorambucil, and morpholino-doxorubicin.
  • benzodiazepines e.g., pyrrolobenzodiazepines and indolinobenzodiazepines; see also Table 1: compounds D1-D10 and DGN462
  • ADCs can be prepared by using a linking group in order to link a drug or prodrug to the antibody or functional equivalent.
  • Suitable linking groups are well known in the art and include, for example, disulfide groups, thioether groups, acid labile groups, photolabile groups, peptidase labile groups and esterase labile groups.
  • the drug or prodrug may, for example, be linked to the antibody or fragment thereof through a disulfide bond.
  • the linker molecule or crosslinking agent can include a reactive chemical group that can react with the antibody or fragment thereof.
  • the reactive chemical groups for reaction with the cell-binding agent can be, for example, N-succinimidyl esters and N-sulfosuccinimidyl esters.
  • the linker molecule comprises a reactive chemical group, such as a dithiopyridyl group that reacts with the drug to form a disulfide bond.
  • Linker molecules include, for example, N-succinmidyl 3-(2-pyridyldithio) propionate (SPDP) (see, e.g., Carlsson et al., Biochem, J., 173:723-737 (1978)), N-succinimidyl 4-(2-pyridyldithio)butanoate (SPDB) (see, e.g., U.S. Pat. No. 4,563,304), N-succinimidyl 4-(2-pyridyldithio)2-sulfobutanoate (sulfo-SPDB) (see US Publication No.
  • SPDP N-succinmidyl 3-(2-pyridyldithio) propionate
  • SPDB N-succinimidyl 4-(2-pyridyldithio)butanoate
  • sulfo-SPDB see US Publication No.
  • N-succinimidyl 4-(2-pyridyldithio)pentanoate (SPP) see, e.g., CAS Registry number 341498-08-6
  • 2-iminothiolane or acetylsuccinic anhydride
  • succinimidyl 4-[N-maleimidomethyl]cyclohexane-carboxylate (SMCC) succinimidyl 4-[N-maleimidomethyl]cyclohexane-carboxylate
  • the antibody or cell binding agent can be modified with crosslinking reagents and the antibody or cell binding agent containing free or protected thiol groups thus derived is then reacted with a disulfide- or thiol-containing maytansinoid to produce conjugates.
  • the conjugates can be purified by chromatography, including but not limited to HPLC, size-exclusion, adsorption, ion exchange and affinity capture, dialysis or tangential flow filtration.
  • an antibody is linked to cytotoxic drugs via disulfide bonds and a polyethylene glycol spacer in enhancing the potency, solubility or the efficacy of the ADC.
  • cleavable hydrophilic linkers are described, for example, in WO2009/0134976.
  • the additional benefit of this linker design is the desired high monomer ratio and the minimal aggregation of the antibody-drug conjugate.
  • conjugates of cell-binding agents and drugs linked via disulfide group (—S—S—) bearing polyethylene glycol spacers ((CH 2 CH 2 O) n 22 1-14 ) with a narrow range of drug load of 2-8 are described that show relatively high potent biological activity toward cancer cells and have the desired biochemical properties of high conjugation yield and high monomer ratio with minimal protein aggregation.
  • the present invention includes aspects wherein about 2 to about 8 drug molecules, for example, maytansinoids, benzodiazepine compounds, auristatins, DNA alkylators, or other compounds of interest, are linked to an antibody or fragment thereof, the anti-tumor effect of the conjugate is much more efficacious as compared to a drug load of a lesser or higher number of drugs linked to the same cell binding agent.
  • drug molecules for example, maytansinoids, benzodiazepine compounds, auristatins, DNA alkylators, or other compounds of interest
  • the drug to antibody ratio averages from about 2 to about 8 (e.g., 1.9, 2.0, 2.1, 2,2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3,6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1).
  • about 2 to about 8 e.g., 1.9, 2.0, 2.1, 2,2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3,
  • cytotoxic agents useful in the present invention are maytansinoids and maytansinoid analogs.
  • suitable maytansinoids include esters of maytansinol and maytansinoid analogs. Included are any drugs that inhibit microtubule formation and that are highly toxic to mammalian cells, as are maytansinol and maytansinol analogs.
  • an ADC of the invention comprises a maytansinoid.
  • Maytansinoids useful in the invention include, but are not limited to, N 2′ -deacetyl-N 2′ -(3-mercapto-1-oxopropyl)-maytansine (DM1), N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansinoid (termed DM3), and N 2′ -deacetyl-N 2′ -(4-mercapto-4-methyl-1-oxopentyl) maytansine (DM4).
  • DM1 N 2′ -deacetyl-N 2′ -(3-mercapto-1-oxopropyl)-maytansine
  • DM3 N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansinoid
  • DM4 N 2
  • DM1 is represented by the following structural formula:
  • DM4 is represented by the following structural formula:
  • suitable maytansinol esters include those having a modified aromatic ring And those having modifications at other positions.
  • Such suitable maytansinoids are disclosed in U.S. Pat. Nos. 4,424,219; 4,256,746; 4,294,757; 4,307,016; 4,313,946; 4,315,929; 4,331,598; 4,361,650; 4,362,663; 4,364,866; 4,450,254; 4,322,348; 4,371,533; 5,208,020; 5,416,064; 5,475,092; 5,585,499; 5,846,545; 6,333,410; 7,276,497 and 7,473,796.
  • N 2′ -deacetyl-N 2′ (4-mercapato-1-oxopentyl)-maytansine (termed DM3), represented by the following structural formula (V):
  • positions on maytansinoids can serve as the position to chemically link the linking moiety.
  • the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with hydroxy and the C-20 position having a hydroxy group are all expected to be useful.
  • the C-3 position serves as the position to chemically link the linking moiety
  • the C-3 position of maytansinol serves as the position to chemically link the linking moiety.
  • a solution of an antibody in aqueous butter can be incubated with a molar excess of maytansinoids having a disulfide moiety that bears a reactive group.
  • the reaction mixture can be quenched by addition of excess amine (such as ethanolamine, taurine, etc.).
  • excess amine such as ethanolamine, taurine, etc.
  • the maytansinoid-antibody conjugate can then be purified by gel filtration.
  • the average number of maytansinoid molecules bound per antibody molecule can be determined by measuring spectrophotometrically the absorbance at 252 nm and 280 nm and determining the molar concentration of the antibody and the molar concentration of the drug. An exemplary calculation is shown herein below for huMov19-sulfo-SPDB-DM4. The average number of maytansinoid molecules per antibody is then calculated by dividing the molar concentration of the drug by the molar concentration of the antibody.
  • the average number of maytansinoid molecules/antibody can be, for example, 1-10 or 2-5. In some embodiments, the average number of maytansinoid molecules/antibody is 3.4.
  • an ADC of the invention comprises a benzodiazepine.
  • Benzodiazepines useful in the invention include, for example, pyrrolobenzodiazepines and indolinobenzodiazepines (see also Table 1: compounds D1-D10 and DGN462).
  • the benzodiazepine compounds are selected from the representative cytotoxic agents D1-D10 and DGN462 listed In Table 1.
  • DGN462 is described, for example, in U.S. Pat. No. 8,765,740, which is incorporated herein by reference in its entirety.
  • Compound D2 is described, for example, in U.S. Provisional Appl. Ser.
  • the present invention further provides pharmaceutical compositions comprising one or more of the ADCs described herein.
  • the pharmaceutical compositions further comprise a pharmaceutically acceptable vehicle. These pharmaceutical compositions find use in inhibiting tumor growth and treating cancer in human patients.
  • Exemplary antibody drug conjugates used in the pharmaceutical compositions of the invention include, without limitation: huMov19-sulfo-SPDB-DM4, huMov19-sulfo-SPDB-D1, huMov19-D2, huMov19-sulfo-SPDB-D10, huMov19sulfo-SPDB-DGN462, huMy9-6-sulfo-SPDB-D1, huMy9-6-D2, huMy9-6-sulfo-SPDB-D10, huMy9-6-sulfo-SPDB-DGN462, huAnti-CD123-sulfo-SPDB-D1, huAnti-CD123-D2, huAnti-CD123-sulfo-SPDB-D10, huAnti-CD123-sulfo-SPDB-DGN462, huB4-SPDB-DM4, huDS6-SPDB-
  • formulations are prepared for storage and use by combining a purified ADC of the present invention with a pharmaceutically acceptable vehicle (e.g. carrier, excipient) (Remington, The Science and Practice of Pharmacy 20th Edition Mack Publishing, 2000).
  • a pharmaceutically acceptable vehicle e.g. carrier, excipient
  • the invention provides for the formulation of such compositions based on drug concentration.
  • an ADC of the invention is provided in a suitable carrier, diluent and/or excipient, such as 0.9% saline (0.9% w/v NaCl), 5% (w/v) dextrose; and may also contain a stabilizing agent such as Tween 20.
  • the ADC is provided in an IV bag or in a drug vial.
  • Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers, such as phosphate, citrate, acetate, succinate and other organic acids; salts such as sodium chloride antioxidants including ascorbic acid and methionine; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol.
  • nontoxic buffers such as phosphate, citrate, acetate, succinate and other organic acids
  • salts such as sodium chloride antioxidants including ascorbic acid and methionine
  • amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine
  • carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dex
  • compositions of the present invention can be administered in any number of ways for either local or systemic treatment.
  • Administration can be parenteral including intravenous, intra-arterial, or infusion; oral; transdermal; or intracranial (e.g., intrathecal or intraventricular) administration.
  • kits that comprise antibody drug conjugates (ADC) that can be used to perform the methods described herein.
  • ADC antibody drug conjugates
  • a kit comprises an ADC in one or more containers, where the amount of ADC is based on the drug concentration and where the amount of ADC varies by no more than ⁇ 10% from specification.
  • the disclosed ADCs can be readily incorporated into one of the established kit formats that are well known in the art.
  • the kit may include instructions for use of the ADC for patient therapy. The instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
  • the anti-FOLR1 monoclonal antibody moiety of huMov19-sulfo-SPDB-DM4 targets and binds to the cell surface antigen FOLR1 (also known as FR ⁇ ). After antibody-antigen interaction and internalization, the immunoconjugate releases DM4, which binds to tubulin and disrupts microtubule assembly/disassembly dynamics, thereby inhibiting cell division and cell growth of FOLR1-expressing tumor cells.
  • FOLR1 a member of the folate receptor family is overexpressed on a variety of epithelial-derived cancer cells.
  • ADC antibody drug conjugate
  • DAR drug antibody ratio
  • MAR maytansinoid-to-antibody ratio
  • the cytotoxic potency When the conjugate concentration is diluted to half the concentration of the reference standard, the cytotoxic potency is half that of the reference standard. Likewise when the starting concentration of the conjugate is double that of the reference standard, the cytotoxic potency is double relative to the reference standard. Details of the Specific Cytotoxicity Assay are provided herein below in Example 6.
  • huMov19-sulfo-SPDB-DM4 it is desirable to achieve as high an ADC level as possible to achieve efficacy without approaching the ocular toxicity threshold.
  • FIG. 3 when huMov19-sulfo-SPDB-DM4 DAR is approximately 3.4, and dosage ranges between 3.3 and 7 mg/kg, 32% of patients were found to be above the ocular toxicity level.
  • huMov19-sulfo-SPDB-DM4 DAR is 2.9, and dosage ranges between 3.3 and 7 mg/kg, 13% of patients would be expected to be above the ocular toxicity level.
  • huMov19-sulfo-SPDB-DM4 DAR When huMov19-sulfo-SPDB-DM4 DAR is 3.4, and dosage ranges between 3.3 and 7 mg/kg, 32% of patients are expected to be above the ocular toxicity level based on the decreased DM4 dose they would receive. When huMov19-sulfo-SPDB-DM4 DAR is 3.9, and dosage ranges between 3.3 and 7 mg/kg, 48% of patients are expected to be above the ocular toxicity level based on the increased DM4 dose they would receive.
  • FIG. 4 demonstrates the importance of ensuring that patients receive a dose within a narrow intended range. Ideally, to ensure maximum efficacy and safety, patients would receive a dose of huMov19-sulfo-SPDB-DM4 that approaches, hut that does not exceed, the ocular toxicity threshold.
  • toxicity depends on the amount of DM4 administered.
  • Preclinical efficacy studies with huMov19-sulfo-SPDB-DM4 showed that there is no DAR dependency when DM4 dose is the same.
  • preclinical toxicity studies with huMov19-sulfo-SPDB-DM4 and many other conjugates showed that toxicity is driven by linked DM4 dose regardless of DAR.
  • the KB cell line was established from a HeLa cell contamination of a tumor cell. It is used as a tumor model because it forms a tumor in nude mice with reproducible characteristics and over expresses the folate receptor.
  • the IGROV-1 tumor model is derived from a human ovarian carcinoma.
  • HuMov19-sulfo-SPDB-DM4 with different DARs were administered to mice bearing KB or IGROV-1 tumor xenografts, at a DM4 dose of 25 ⁇ g/kg and variable antibody dosages.
  • DM4 dose 25 ⁇ g/kg and variable antibody dosages.
  • FIG. 5 as long as the same DM4 dose was administered, the DAR did not impact efficacy.
  • the ADC huEGFR-7R-SMCC-DM1 was administered at a fixed DM1 dose of 3.0 mg/kg.
  • the DAR varied (e.g., 2.3, 3.5, 6.3, 10.1), but DM1 dosage was held constant.
  • Mean body weight (BW) change was monitored as an indicator of toxicity. The body weight loss was similar for the different DAR conjugates indicating that toxicity was independent of DAR as long as the DM1 dose was held constant ( FIG. 7 ).
  • the ADC huDS6-SPDB-DM4 (also known as “huDS6-DM4”) is the humanized monoclonal antibody, huDS6, linked to DM4, a potent cytotoxic maytansinoid through a cleavable disulfide cross-linking agent N-Succinimidyl-4-2-pyridyldithio butanoate acid (SPDB).
  • the ADC huDS6-SPDB-DM4 targets solid tumors such as ovarian, breast, cervical, lung and pancreatic carcinomas.
  • the ADC huB4-SPDB-DM4 (also known as “huB4-DM4”) is a novel antibody-drug conjugate that is composed of a humanized monoclonal IgG1 anti-CD19 antibody (huB4) attached to DM4 through a cleavable disulfide cross-linking agent N-Succinimidyl-4-2-pyridyldithio butanoate acid (SPDB).
  • the ADC huMy9-6-SPDB-DM4 (also known as “huMy9-6-DM4”) is an ADC that specifically binds CD33, a siglec family antigen expressed primarily on myeloid cells.
  • the ADC huMy9-6-SPDB-DM4 has undergone clinical evaluation for the treatment of acute myeloid leukemia.
  • the specified conjugate was administered at 3-4 different doses and mouse survival was measured as an indicator of toxicity.
  • the DAR range for the 4 conjugates was narrow (ranging from 3.49 to 4.0); thus, the LD 50 range (the dose that is lethal to 50% of the animals was also narrow (DM4 dose between 1.6-2 mg/kg).
  • Conjugates having different DAR were administered at 80 mg/kg antibody dose, and mouse survival was measured as an indicator of toxicity ( FIG. 8 ).
  • the similar result for all conjugates, regardless of DAR, indicates that the toxicity is driven by the total DM4 dose administered. That is, within the DAR range of 2.1 to 5.1 the toxicity is not affected by different DAR.
  • FIG. 9A shows the variability inherent in formulating antibody drug conjugates based on antibody concentration, even, when remaining within the allowable ranges of the specification.
  • the concentration of antibody is measured, and the antibody is diluted to reach the target antibody concentration, which is 5.0 mg/ml for huMov19-sulfo-SPDB-DM4.
  • the target antibody concentration 5.0 mg/ml
  • the target DAR 3.4
  • the DM4 concentration is 91.1 ⁇ g/ml.
  • the antibody concentration in the finished drug product is allowed to vary from the target concentration.
  • the antibody concentration in the finished product could be as low as 4.0 mg/ml or as high as 6.0 mg/ml.
  • the DM4 concentration in the finished drug product could be as low as 62.1 ⁇ g/ml or as high as 125.4 ⁇ g/ml.
  • Formulating the ADC composition based on DM4 concentration and adding a DM4concentration specification of ⁇ 10 % significantly narrows the potency present in the finished drug product to ⁇ 10% of the target DM4 concentration (shown in highlight; ⁇ 10% specification).
  • FIG. 9B The liability for batches failing to conform to specification is shown at FIG. 9B where the target DM4 concentration and DAR are highlighted. DM4 concentration is shown at the top, DAR is shown at left, and the resulting antibody concentration is shown within the highlighted box (5.0 mg/ml). When the antibody concentration varies more than ⁇ 20% from the target, that batch is out of specification. The concentration of antibody present in batches failing to conform to specification is shown in bold ( FIG. 9B ). The risk of batches falling to conform to specification is fairly low.
  • the current formulation specification allows ⁇ 20% variation in antibody-based concentration (4.0-6.0 mg/mL).
  • the ADC potency could vary by as much as ⁇ 35% depending on the DAR.
  • potency is permitted to vary by only ⁇ 10%.
  • the new formulation method eliminates DAR potency dependence by formulating to a narrow range of DM4 concentration. Such a formulation strategy only slightly increases the risk of a batch failing to conform to specification due to the antibody concentration being outside its specification.
  • the ADC huMov19-sulfo-SPDB-DM4 which comprises an huMov19 antibody, SPDB linker and the cytotoxic drug, DM4, is an example of an ADC where the in vitro potency, in vivo efficacy, and in vivo, toxicity are independent of DAR and are driven entirely by the administered concentration of DM4.
  • huMov19-sulfo-SPDB-DM4 is a good candidate for formulating by DM4 rather than huMov19 concentration.
  • a series of huMov19-sulfo-SPDB--DM4conjugates having a range of DARs were manufactured.
  • the conjugates were purified into base formulation buffer (10 mM sodium acetate, 9% (w/v) sucrose, pH 5.0) and the DM4 and huMov19 antibody concentration for each sample was measured spectrophotometrically at wavelengths 252 nm and 280 nm, respectively.
  • the molar concentration of DM4 and huMov19 antibody comprising the conjugate is calculated as follows:
  • Each of the various DAR conjugates was formulated in two different ways: one was diluted with the base formulation buffer to reach the target huMov19 antibody concentrations within the specification range of 5.0 mg/mL ⁇ 20%.
  • the various DAR conjugates were formulated to target DM4 concentrations within the proposed specification of 91.1 ⁇ g/mL ⁇ 10%. All samples were subjected to Specific Cytotoxicity assay.
  • the Specific Cytotoxicity assay involves incubating Folate Receptor 1 (FOLR1) positive cells (KB) in the presence of media containing a dilution series of huMov19-sulfo-SPDB-DM4drug conjugate in duplicate wells of a sterile, 96-well, flat bottomed black tissue culture plate with clear bottom.
  • Each assay plate contains a reference, control and a test article series of identical dilutions in wells with KB cells and media blanks. After a 4-day incubation period at 37° C. ⁇ 2° C., the plates are removed from the incubator and allowed to equilibrate to room temperature for 1 hour prior to the addition of CellTiter-GloTM Luminescent Cell Viability Reagent.
  • the plates are incubated for an additional 2 hours prior to analyzing for and recording the luminescent signal on the Victor III plate reader.
  • CellTiter-GloTM uses a unique, stable form of luciferase to measure ATP as an indicator of viable cells.
  • the luminescent signal produced is directly proportional to the number of viable cells present in the well and likewise inversely proportional to the cytotoxicity of the drug in that well. Because the luciferase reaction requires ATP, conditions have been created such that the amount of light produced is proportional to the amount of ATP present, reflecting the number of viable cells.
  • the three plate data file is imported into PLA 2.0 software and the EC 50 values for reference and test article are determined from the constrained 4 parameter logistic curve fit using all 6 replicates for each sample. For samples passing acceptance criteria for slope difference and parallelism, the % relative potency of the test article is reported from the IC50s derived from the constrained 4PL curve fit. Percent potency is calculated as follows.
  • % ⁇ ⁇ Potency EC 50 ( reference ⁇ ⁇ standard ) ⁇ ⁇ 100 ⁇ % EC 50 ( test ⁇ ⁇ article )
  • test article EC 50 is lower than the reference standard EC 50 , this indicates that the test article has greater potency than the reference standard and the calculated % potency will be greater than 100%. Conversely, if the test article EC 50 is greater than the reference standard EC 50 this indicates that the test article has less potency than the reference standard and the calculated % potency will be less than 100%.
  • Targeting Variable Antibody and Drug Concentration Tightens the Specification Window over a Broader DAR Range
  • the targeted drug concentration may be desirable to allow variation in the targeted drug concentration to arrive at smaller variations for both drug and antibody concentrations rather than large variations in the non-targeted concentration (e.g., the antibody concentration).
  • Formulating an ADC composition using such method maximizes specification range by targeting the middle of the range where both the antibody and drug concentration overlap at a particular DAR value.
  • drug specifications are tighter than the antibody specifications (e.g. ⁇ 10% for drug versus ⁇ 15% for the antibody).
  • allowing smaller variations in antibody and drug concentrations provide an additional control strategy to achieve tighter drug concentration specification (rather than an absolute target) while minimizing risk of batches that, although not conforming to specification, would be perfectly safe to use.
  • huMy9-6-sulfo-SPDB-DGN462 which is a CD33-targeted antibody drug conjugate comprising the antibody huMy9-6, conjugated to a novel DNA-alkylating agent, DGN462 via a cleavable disulfide linker, sulfo-SPDB.
  • the upper and lower limit of the antibody specification range is indicated by a dashed line
  • the X axis indicates the DAR of a batch of drug, where the target DAR is 2.7
  • the vertical lines show the upper and lower DGN462 specification limits at a given DAR
  • the dark gray diamond on each vertical line indicates the overlap between the DGN462 specification range and the antibody specification range at a particular DAR.
  • FIG. 11A when DAR for the batch is 2.7, the center of the DGN462 specification range falls squarely within the center of the antibody range.
  • FIG. 11A when DAR for the batch is 2.7, the center of the DGN462 specification range falls squarely within the center of the antibody range.
  • FIGS. 11A-11C show the improvement to be gained by allowing both DGN462 and antibody concentration to vary—particularly when the DAR approaches the upper and lower DAR specification limits (e.g., 3.0-3.1 and 2.3-2.4).
  • Targeting a variable drug concentration identifies the middle of the range where the antibody concentration specification range and the drug concentration specification range overlap ( FIG. 11A ). The center of the effective range of total drug is well below the antibody upper specification limit.
  • the method of targeting a variable DGD462 limits the target antibody concentration to vary ⁇ 10% from target instead of the full ⁇ 15% generating a more consistent product.
  • FIG. 11C illustrates the improvement provided by the method of targeting a variable drug concentration at the upper and lower DAR specification limits, where the DGN462 concentration and the antibody concentration are maintained between narrower limits of 4% (open ovals) compared to where the antibody concentration is used to achieve a target drug concentration (gray diamonds), in which the fixed target total drug exceeds the upper specification limit for antibody at a DAR of 2.3.
  • Table 4 illustrates methods used to calculate the upper and lower specification limit of DGN462 (USL DGN462LSL DGN462).
  • Upper Antibody Concentration Specification Limit is Constant.
  • DAR is empirically determined for a batch of antibody drug conjugate. The calculations according to the desired upper and lower specification limits were performed as follows:
  • the values “2.30” and “1.70” define the upper end lower antibody specification limits.
  • the denominator is the molecular weight of the antibody.
  • an ADC is formulated by targeting variable drug concentration.
  • the ADC includes non-functional antibody, huMov19 (MW of 145676 g/mol) conjugated to D2 (MW 961.05 g/mol) with a target DAR of 2.7, antibody concentration of 2.0 mg/mL, and cytotoxic agent concentration of 39.2 ⁇ g/mL.
  • the ADC is formulated to target a variable concentration of drug to minimize the offset from target for the antibody concentration.
  • FIG. 11A targeting a variable drug concentration varies the resulting antibody concentration by ⁇ 10% (1.8-2.2) versus ⁇ 15% when a static drug concentration is utilized ( FIG. 11A ; see, e.g., Tables 4-7).
  • Targeting a variable drug identifies the middle of the range where the antibody specification range and the drug specification range overlap ( FIG. 11A ; see, e.g., Tables 4-7).
  • Table 5 below illustrates methods used to calculate the upper and lower specification limit of D2 (USL D2, LSL D2).
  • the Upper and Lower Antibody Concentration Specification Limits are constant and the DAR is empirically determined.
  • an ADC is formulated by targeting variable cytotoxic agent concentration.
  • the ADC includes a functional antibody, huEGFR-7R (MW of 144975 g/mol) corrugated to D1 (MW 838 g/mol) with a target DAR of 2.7, antibody concentration of 2.0 mg/mL, and cytotoxic agent concentration of 34.3 ⁇ g/mL.
  • the ADC is formulated to target a variable concentration of cytotoxic agent to minimize the offset from target for the antibody concentration.
  • Table 6 below illustrates methods used to calculate the upper and lower specification limit of D1 (USL D1, LSL D1).
  • the Upper and Lower Antibody Concentration Specification Limits are constant and the DAR is empirically determined.
  • an ADC is formulated by targeting variable cytotoxic agent concentration.
  • the ADC includes a functional antibody, huMy9-6 (MW of 146192 g/mol) conjugated to D10 (MW 1062.22 g/mol) with a target DAR of 2.7, antibody concentration of 2.0 mg/mL, and cytotoxic agent concentration of 44.0 ⁇ g/mL.
  • the ADC is formulated to target a variable concentration of cytotoxic agent to minimize the offset from target for the antibody concentration.
  • Table 7 below illustrates methods used to calculate the upper and lower specification limit of D1 (USL D1, LSL D1).
  • the Upper and Lower Antibody Concentration Specification Limits are constant and the DAR is empirically determined.
US14/843,769 2014-09-02 2015-09-02 Methods for formulating antibody drug conjugate compositions Abandoned US20160058884A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/843,769 US20160058884A1 (en) 2014-09-02 2015-09-02 Methods for formulating antibody drug conjugate compositions
US15/678,505 US10603388B2 (en) 2014-09-02 2017-08-16 Methods for formulating antibody drug conjugate compositions
US16/407,384 US20190365917A1 (en) 2014-09-02 2019-05-09 Methods for formulating antibody drug conjugate compositions
US18/146,307 US20230364252A1 (en) 2014-09-02 2022-12-23 Methods for formulating antibody drug conjugate compositions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462044592P 2014-09-02 2014-09-02
US14/843,769 US20160058884A1 (en) 2014-09-02 2015-09-02 Methods for formulating antibody drug conjugate compositions

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/678,505 Division US10603388B2 (en) 2014-09-02 2017-08-16 Methods for formulating antibody drug conjugate compositions
US16/407,384 Division US20190365917A1 (en) 2014-09-02 2019-05-09 Methods for formulating antibody drug conjugate compositions

Publications (1)

Publication Number Publication Date
US20160058884A1 true US20160058884A1 (en) 2016-03-03

Family

ID=55401285

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/843,769 Abandoned US20160058884A1 (en) 2014-09-02 2015-09-02 Methods for formulating antibody drug conjugate compositions
US15/678,505 Expired - Fee Related US10603388B2 (en) 2014-09-02 2017-08-16 Methods for formulating antibody drug conjugate compositions
US16/407,384 Abandoned US20190365917A1 (en) 2014-09-02 2019-05-09 Methods for formulating antibody drug conjugate compositions
US18/146,307 Pending US20230364252A1 (en) 2014-09-02 2022-12-23 Methods for formulating antibody drug conjugate compositions

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/678,505 Expired - Fee Related US10603388B2 (en) 2014-09-02 2017-08-16 Methods for formulating antibody drug conjugate compositions
US16/407,384 Abandoned US20190365917A1 (en) 2014-09-02 2019-05-09 Methods for formulating antibody drug conjugate compositions
US18/146,307 Pending US20230364252A1 (en) 2014-09-02 2022-12-23 Methods for formulating antibody drug conjugate compositions

Country Status (13)

Country Link
US (4) US20160058884A1 (ja)
EP (2) EP3188761A4 (ja)
JP (4) JP2017526682A (ja)
KR (2) KR20240011258A (ja)
CN (3) CN109147874A (ja)
AU (2) AU2015311951B2 (ja)
CA (1) CA2958882A1 (ja)
HK (1) HK1254612A1 (ja)
IL (2) IL250768B (ja)
MA (2) MA40539A (ja)
RU (1) RU2741470C9 (ja)
SG (1) SG11201701328XA (ja)
WO (1) WO2016036861A1 (ja)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018085359A1 (en) * 2016-11-02 2018-05-11 Immunogen, Inc. Combination treatment with antibody-drug conjugates and parp inhibitors
US10603388B2 (en) 2014-09-02 2020-03-31 Immunogen, Inc. Methods for formulating antibody drug conjugate compositions
US11932701B2 (en) 2013-08-30 2024-03-19 Immunogen, Inc. Method for increasing the efficacy of cancer therapy by administering an anti-FOLR1 immunoconjugate

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG193514A1 (en) 2011-04-01 2013-10-30 Immunogen Inc Methods for increasing efficacy of folr1 cancer therapy
MD3313845T2 (ro) * 2015-06-29 2021-02-28 Immunogen Inc Conjugați cu anticorpi având cisteină modificată prin inginerie genetică
EP3463477A4 (en) 2016-06-07 2020-03-04 NanoPharmaceuticals LLC NON-CLeavable POLYMER CONJUGATED WITH THYROID ANTAGONISTS OF avß3 INTEGRIN
CA3044391A1 (en) 2016-11-23 2018-05-31 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
CN111587125A (zh) * 2018-01-12 2020-08-25 伊缪诺金公司 抗体药物缀合、纯化和调配的方法
US10328043B1 (en) 2018-04-11 2019-06-25 Nanopharmaceuticals, Llc. Composition and method for dual targeting in treatment of neuroendocrine tumors
US11351137B2 (en) 2018-04-11 2022-06-07 Nanopharmaceuticals Llc Composition and method for dual targeting in treatment of neuroendocrine tumors
EP3924378A4 (en) 2019-02-15 2023-04-05 WuXi Biologics Ireland Limited METHODS FOR THE PRODUCTION OF ANTIBODY-DRUG CONJUGATES WITH IMPROVED HOMOGENEITY
WO2020223221A1 (en) 2019-04-29 2020-11-05 Immunogen, Inc. Biparatopic fr-alpha antibodies and immunoconjugates
US10961204B1 (en) 2020-04-29 2021-03-30 Nanopharmaceuticals Llc Composition of scalable thyrointegrin antagonists with improved blood brain barrier penetration and retention into brain tumors
US11723888B2 (en) 2021-12-09 2023-08-15 Nanopharmaceuticals Llc Polymer conjugated thyrointegrin antagonists

Family Cites Families (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
JPS55102583A (en) 1979-01-31 1980-08-05 Takeda Chem Ind Ltd 20-acyloxy-20-demethylmaytansinoid compound
JPS55162791A (en) 1979-06-05 1980-12-18 Takeda Chem Ind Ltd Antibiotic c-15003pnd and its preparation
JPS5645483A (en) 1979-09-19 1981-04-25 Takeda Chem Ind Ltd C-15003phm and its preparation
JPS5645485A (en) 1979-09-21 1981-04-25 Takeda Chem Ind Ltd Production of c-15003pnd
EP0028683A1 (en) 1979-09-21 1981-05-20 Takeda Chemical Industries, Ltd. Antibiotic C-15003 PHO and production thereof
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
WO1982001188A1 (en) 1980-10-08 1982-04-15 Takeda Chemical Industries Ltd 4,5-deoxymaytansinoide compounds and process for preparing same
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4563304A (en) 1981-02-27 1986-01-07 Pharmacia Fine Chemicals Ab Pyridine compounds modifying proteins, polypeptides or polysaccharides
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
DE69233482T2 (de) 1991-05-17 2006-01-12 Merck & Co., Inc. Verfahren zur Verminderung der Immunogenität der variablen Antikörperdomänen
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
CA2076465C (en) 1992-03-25 2002-11-26 Ravi V. J. Chari Cell binding agent conjugates of analogues and derivatives of cc-1065
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
CA2219361C (en) 1995-04-27 2012-02-28 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc HUMAN ANTIBODIES DERIVED FROM IMMUNIZED XENO MOUSES
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
EP1500329B1 (en) 1996-12-03 2012-03-21 Amgen Fremont Inc. Human antibodies that specifically bind human TNF alpha
CN100387621C (zh) 1997-04-14 2008-05-14 麦可麦脱股份公司 抗人抗原受体的新的生产方法及其用途
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US6995145B1 (en) * 1999-06-04 2006-02-07 Au Jessie L-S Methods and compositions for modulating drug activity through telomere damage
US6333410B1 (en) 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
US6716821B2 (en) 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
GB0202319D0 (en) 2002-02-01 2002-03-20 Calex Electronics Ltd Apparatus
KR20050010756A (ko) * 2002-02-22 2005-01-28 뉴 리버 파마슈티칼스, 인크. 피험자 간 약물 혈청 농도의 변동성을 감소시키기 위한펩티드-약물 접합체의 용도
US20090068178A1 (en) * 2002-05-08 2009-03-12 Genentech, Inc. Compositions and Methods for the Treatment of Tumor of Hematopoietic Origin
US20050276812A1 (en) * 2004-06-01 2005-12-15 Genentech, Inc. Antibody-drug conjugates and methods
AU2003285878B2 (en) 2002-11-07 2011-04-28 Immunogen, Inc. Anti-CD33 antibodies and method for treatment of acute myeloid leukemia using the same
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
CN102940889A (zh) * 2003-05-14 2013-02-27 伊缪诺金公司 药物缀合物组合物
EP3851126A1 (en) 2003-05-20 2021-07-21 ImmunoGen, Inc. Maytansinoid-cell-binding agent conjugates
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
US7834155B2 (en) 2003-07-21 2010-11-16 Immunogen Inc. CA6 antigen-specific cytotoxic conjugate and methods of using the same
WO2005009369A2 (en) 2003-07-21 2005-02-03 Immunogen, Inc. A ca6 antigen-specific cytotoxic conjugate and methods of using the same
EA013323B1 (ru) 2004-12-09 2010-04-30 Сентокор, Инк. Иммуноконъюгаты против интегрина, способы и варианты применения
US20110166319A1 (en) * 2005-02-11 2011-07-07 Immunogen, Inc. Process for preparing purified drug conjugates
WO2007002422A1 (en) 2005-06-23 2007-01-04 Georgia Tech Research Corporation Systems and methods for integrated plasma processing of waste
CA2615761A1 (en) 2005-08-22 2007-03-01 Immunogen, Inc. Humanized anti-ca6 antibodies and methods of using the same
ES2533992T3 (es) * 2005-08-24 2015-04-16 Immunogen, Inc. Procedimiento para preparar conjugados de anticuerpo maitansinoide
PT1813614E (pt) * 2006-01-25 2012-01-09 Sanofi Sa Agentes citotóxicos compreendendo novos derivados de tomaimicina
CA2638811A1 (en) * 2006-02-03 2007-08-16 Medimmune, Llc Protein formulations
WO2007140371A2 (en) * 2006-05-30 2007-12-06 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
WO2008119353A1 (en) 2007-03-29 2008-10-09 Genmab A/S Bispecific antibodies and methods for production thereof
US20090163698A1 (en) * 2007-05-11 2009-06-25 John Joseph Grigsby Method for Preparing Antibody Conjugates
US7576505B2 (en) * 2007-06-21 2009-08-18 Jack Chen Device for finding a home position for a moveable member
US20090208979A1 (en) * 2008-02-14 2009-08-20 Technion Research And Development Foundation Ltd. Method for identifying antipsychotic drug candidates
EP2276506A4 (en) 2008-04-30 2014-05-07 Immunogen Inc EFFICIENT CONJUGATES AND HYDROPHILIC BINDER
KR20230003298A (ko) 2008-04-30 2023-01-05 이뮤노젠 아이엔씨 가교제 및 그 용도
KR101725170B1 (ko) * 2009-05-06 2017-04-10 바이오테스트 아게 Cd138을 표적으로 하는 면역접합체의 용도
AR078470A1 (es) * 2009-10-02 2011-11-09 Sanofi Aventis Anticuerpos que se unen especificamente al receptor epha2
SG10201501342UA (en) * 2010-02-24 2015-04-29 Immunogen Inc Folate receptor 1 antibodies and immunoconjugates and uses thereof
MX338932B (es) 2010-03-12 2016-05-06 Immunogen Inc Moleculas de union cd37 y sus inmunoconjugados.
SG184427A1 (en) 2010-04-20 2012-11-29 Genmab As Heterodimeric antibody fc-containing proteins and methods for production thereof
EA201390472A1 (ru) 2010-10-29 2014-02-28 Иммьюноджен, Инк. Новые молекулы, связывающиеся с egfr, и их иммуноконъюгаты
RU2748733C2 (ru) * 2011-02-15 2021-05-31 Иммуноджен, Инк. Цитотоксические производные бензодиазепина
MX2013011385A (es) * 2011-04-01 2014-03-13 Immunogen Inc Moleculas de union a cd37 y sus inmunoconjugados.
SG193514A1 (en) 2011-04-01 2013-10-30 Immunogen Inc Methods for increasing efficacy of folr1 cancer therapy
WO2012138749A1 (en) 2011-04-04 2012-10-11 Immunogen, Inc. Methods for decreasing ocular toxicity of antibody drug conjugates
US9289512B2 (en) 2011-06-21 2016-03-22 Immunogen, Inc. Maytansinoid derivatives with peptide linker and conjugates thereof
EP2550975A1 (en) * 2011-07-29 2013-01-30 Sanofi Combination therapy for the treatment of CD19+ B-cell malignancies symptoms comprising an anti-CD19 maytansinoid immunoconjugate and rituximab
KR102007055B1 (ko) 2011-09-22 2019-08-02 암젠 인크 Cd27l 항원 결합 단백질
CA2856411A1 (en) 2011-11-21 2013-05-30 Immunogen, Inc. Method of treatment of tumors that are resistant to egfr therapies by egfr antibody cytotoxic agent conjugate
RU2661083C2 (ru) * 2012-10-04 2018-07-11 Иммуноджен, Инк. Использование пвдф-мембраны для очистки конъюгатов клеточно-связывающий агент - цитотоксический агент
PT2922875T (pt) * 2012-11-20 2017-05-31 Sanofi Sa Anticorpos anti-ceacam5 e suas utilizações
SI2958944T1 (sl) * 2013-02-22 2019-08-30 Abb Vie Stemcentrx Llc Konjugati protiDLL3-protitelo-PBD in njihove uporabe
JP2016525560A (ja) * 2013-08-02 2016-08-25 サノフイ 固形腫瘍を処置するための抗−Muc1メイタンシノイドイムノコンジュゲート抗体の使用
WO2016036861A1 (en) 2014-09-02 2016-03-10 Immunogen, Inc. Methods for formulating antibody drug conjugate compositions
WO2016036804A1 (en) * 2014-09-03 2016-03-10 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
TWI697493B (zh) * 2014-09-03 2020-07-01 美商免疫原公司 細胞毒性苯并二氮呯衍生物
TW201609152A (zh) * 2014-09-03 2016-03-16 免疫原公司 包含細胞結合劑及細胞毒性劑之偶聯物
JP2016052860A (ja) 2014-09-04 2016-04-14 株式会社東海理化電機製作所 タイヤ位置登録システム

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11932701B2 (en) 2013-08-30 2024-03-19 Immunogen, Inc. Method for increasing the efficacy of cancer therapy by administering an anti-FOLR1 immunoconjugate
US10603388B2 (en) 2014-09-02 2020-03-31 Immunogen, Inc. Methods for formulating antibody drug conjugate compositions
WO2018085359A1 (en) * 2016-11-02 2018-05-11 Immunogen, Inc. Combination treatment with antibody-drug conjugates and parp inhibitors

Also Published As

Publication number Publication date
JP2017526682A (ja) 2017-09-14
CN106999604B (zh) 2021-08-03
EP3188761A1 (en) 2017-07-12
RU2741470C2 (ru) 2021-01-26
CA2958882A1 (en) 2016-03-10
JP2020172541A (ja) 2020-10-22
EP3311846A1 (en) 2018-04-25
US20170348428A1 (en) 2017-12-07
HK1254612A1 (zh) 2019-07-26
KR102626976B1 (ko) 2024-01-18
AU2015311951A1 (en) 2017-04-20
CN113842468A (zh) 2021-12-28
US20190365917A1 (en) 2019-12-05
AU2021204047A1 (en) 2021-07-08
IL283777A (en) 2021-07-29
MA40539A (fr) 2016-03-10
RU2017107873A3 (ja) 2019-04-10
CN106999604A (zh) 2017-08-01
US20230364252A1 (en) 2023-11-16
RU2741470C9 (ru) 2021-04-27
AU2015311951B2 (en) 2021-04-08
JP2020122012A (ja) 2020-08-13
EP3188761A4 (en) 2018-04-18
RU2017107873A (ru) 2018-10-03
KR20240011258A (ko) 2024-01-25
CN109147874A (zh) 2019-01-04
MA42561A (fr) 2018-04-25
IL250768A0 (en) 2017-08-31
IL250768B (en) 2021-06-30
SG11201701328XA (en) 2017-03-30
KR20170063668A (ko) 2017-06-08
JP2022050688A (ja) 2022-03-30
US10603388B2 (en) 2020-03-31
WO2016036861A1 (en) 2016-03-10

Similar Documents

Publication Publication Date Title
US20230364252A1 (en) Methods for formulating antibody drug conjugate compositions
US20220378694A1 (en) Anti-FOLR1 Immunoconjugate Dosing Regimens
US8309094B2 (en) Antibody-drug conjugates
JP2020063271A (ja) 部位特異的抗体コンジュゲーション方法および組成物
CN109715666B (zh) 癌症治疗方法和使用结合糖基化pd-l1的抗体的组合的疗法
RU2696579C2 (ru) Схемы применения иммуноконъюгата анти-folr1
KR20180129872A (ko) 글리코실화된 pd-l1에 특이적인 이중 기능 항체 및 이의 사용 방법
CN116808198A (zh) 包含抗-ly75抗体的医药组合
US20230256114A1 (en) Novel maytansinoids as adc payloads and their use for the treatment of cancer
CN112236143A (zh) 医药组合
US20230212181A1 (en) Double payload cancer therapeutics
WO2023089314A1 (en) Pharmaceutical combinations
RU2801307C1 (ru) Схемы применения иммуноконъюгата анти-folr1

Legal Events

Date Code Title Description
AS Assignment

Owner name: IMMUNOGEN, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAYNE, GILLIAN;HERBST, ROBERT W.;BRIDGEWATER, JUMA;SIGNING DATES FROM 20150921 TO 20151012;REEL/FRAME:036791/0415

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION