WO2020092474A1 - Methods and compositions for imaging amyloid deposits - Google Patents

Methods and compositions for imaging amyloid deposits Download PDF

Info

Publication number
WO2020092474A1
WO2020092474A1 PCT/US2019/058720 US2019058720W WO2020092474A1 WO 2020092474 A1 WO2020092474 A1 WO 2020092474A1 US 2019058720 W US2019058720 W US 2019058720W WO 2020092474 A1 WO2020092474 A1 WO 2020092474A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
patient
amyloid deposits
chimeric
Prior art date
Application number
PCT/US2019/058720
Other languages
French (fr)
Inventor
Suzanne Lentzsch
Akiva Mintz
Original Assignee
The Trustees Of Columbia University In The City Of New York
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 The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Priority to JP2021523841A priority Critical patent/JP7337922B2/en
Priority to EP19880738.0A priority patent/EP3873932A4/en
Publication of WO2020092474A1 publication Critical patent/WO2020092474A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1018Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against material from animals or humans
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4709Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present disclosure relates to humanized and chimeric (e.g ., mouse-human) antibodies and antigen-binding fragments thereof having a detectable molecule linked or conjugated thereto and to methods of using the same to detect and image amyloid deposits.
  • humanized and chimeric (e.g ., mouse-human) antibodies and antigen-binding fragments thereof having a detectable molecule linked or conjugated thereto and to methods of using the same to detect and image amyloid deposits.
  • Native antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons composed of two identical light chains and two identical heavy chains. Each light chain is linked to a heavy chain by one disulfide bond, while the number of additional disulfide linkages between the heavy chains varies with different antibody isotypes.
  • the simplest isotype is IgG, which comprises just two light chains and two heavy chains, in which the two heavy chains are linked by two disulfide linkages.
  • Each heavy chain has a variable domain (VH) at one end with a number of adjacent constant domains.
  • Each light chain has a variable domain (VL) at one end and a constant domain at its other end.
  • Each variable domain of the light and heavy chain in an antibody comprises three segments called complementarity-determining regions (“CDR”) or hypervariable regions.
  • CDR complementarity-determining regions
  • Light chains are of two major types, k and l, depending on their constant region. Both k and l light chains may combine with any of the different heavy chain types.
  • Amyloid light-chain amyloidosis (AL amyloidosis, AL, or ALA), also called primary amyloidosis, is the most common form of systemic amyloidosis in the United States.
  • the term“amyloidosis” refers to a cluster of diseases which share a common feature, i.e., the extracellular deposition of pathologic insoluble fibrillar proteins in organs and tissues (Rodney, et al. - NEJM, 25:898).
  • Amyloidosis is caused by malfunction of a person’s antibody- producing cells causing production of abnormal protein fibers which aggregate to form insoluble amyloid deposits in organs and tissues.
  • amyloidosis is determined by the nature of the precursor proteins which form the fibril deposit.
  • the fibrils comprise fragments of immunoglobulin light chains and in secondary amyloidosis, the fibrils comprise amyloid A protein.
  • Modern classification of amyloidosis is based on the nature of the precursor plasma proteins which form the fibril deposit.
  • amyloidosis The precursor plasma proteins are diverse and unrelated. Nevertheless, all precursor deposits produce amyloid deposits that share a common typical b-pleated-sheet configuration, which is responsible for the typical staining properties of the fibrillar deposits. The final stage in the development of amyloidosis is the deposit of amyloid fibrils in the organs of the sufferer. Amyloidosis mortality is high, with current five-year survival rates of about 28%.
  • compositions and methods disclosed herein fulfill this need for detecting such presence and location.
  • compositions and methods for detecting the presence, location, and amount of amyloid deposits comprising those resulting from primary (AL) amyloidosis.
  • the disclosed compositions comprise a humanized or chimeric antibody or fragment thereof (“antigen-binding fragment”) that specifically binds to amyloid deposits (e.g ., amyloid light chain fibrils) and to which is linked a detectable molecule.
  • the disclosed method comprises administering to a subject suspected of having amyloid deposits the above-described composition and detecting the presence, quantity, and/or location of the amyloid deposits by detection of the detectable molecule by diagnostic imaging.
  • the present methods also include a method of stratifying patients for therapy with the disclosed humanized or chimeric antibody based on the affinity of the diagnostic composition for the amyloid deposits.
  • the present methods also include determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy based on the affinity of the diagnostic compositions for the amyloid deposits. That is, one may determine the appropriate dosing of a patient based on the detected affinity (uptake) of the labeled antibody or antibody fragment.
  • a patient showing strong affinity for the labeled antibody or antigen-binding antibody fragment may require a smaller amount of therapeutic antibody or antigen-binding antibody fragment than a patient showing a weaker affinity for the labeled antibody or antigen-binding antibody fragment.
  • the present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the affinity of the labeled antibody or antibody fragment for the amyloid deposits, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment based on the strength of the affinity.
  • the disclosed antibody in the diagnostic compositions comprises a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
  • the antibody comprises a constant region derived from a human lgG1.
  • the antibody binds to amyloid fibrils with a higher affinity than its murine equivalent.
  • the antibody binds to an epitope expressed by the b-pleated sheet configuration of amyloid fibrils with higher affinity than a mouse antibody comprising a VK region of SEQ ID NO: 36 and a VH region of SEQ ID NO: 35.
  • the antibody binds to kappa and lambda amyloid fibrils in vivo.
  • compositions comprising the disclosed humanized or chimeric antibody linked to a detectable label or molecule and a pharmaceutically acceptable carrier.
  • a chimeric antibody useful in the subject methods and compositions may be produced by co-transfection in mammalian cells of the vector constructs 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200 or transfection in mammalian cells of the supervector construct pG1 KD200-11-1 F4.
  • the co-transfection of the vector constructs 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200 or transfection of a supervector construct pG1 KD200- 11-1 F4 takes place in COS cells.
  • the antibody thus produced is designated “chimeric 11-1 F4 antibody”.
  • fragments of the chimeric 11-1 F4 antibody are also useful in the subject methods and compositions.
  • the amyloid deposits detected result from primary amyloidosis.
  • the primary amyloidosis comprises involvement of at least one organ or tissue selected from the group consisting of heart, kidneys, liver, lung, gastrointestinal tract, nervous system, muscular skeletal system, soft tissue, and skin and the deposits are detected in one or more of those organs.
  • the present disclosure provides methods of detecting or imaging amyloid deposits comprising administering to a patient diagnosed with or suspected of having an amyloid deposition disease a diagnostically effective amount of a humanized or chimeric antibody or an antigen-binding fragment thereof having a detectable label or marker linked thereto and detecting the presence, location, and/or quantity of the detectable label bound to amyloid deposits in the patient’s body by diagnostic imaging.
  • the antibody or antigen-binding fragment may comprise: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 52; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51.
  • VH variable heavy chain
  • CDR complementarity determining region
  • VK variable light chain
  • the present disclosure provides compositions of the aforementioned antibody or antibody fragment linked to a detectable molecule and a pharmaceutically-acceptable carrier.
  • the antibody or antigen-binding fragment thereof may be a humanized antibody, while in some embodiments, the antibody or antigen-binding fragment thereof may be a chimeric antibody.
  • the VK region of the antibody or antigen-binding fragment may comprise SEQ ID NO: 47 and the VH region may comprise SEQ ID NO: 48.
  • the antibody or antigen-binding fragment may comprise a constant region that is derived from a human lgG1.
  • the antibody may be chimeric 11 -1 F4 antibody.
  • the present disclosure provides methods of detection of an amyloid deposition disease in a patient suspected of having such disease by administering a labeled antibody or an antigen-binding fragment thereof and detecting the presence of the label in the patient by diagnostic imaging.
  • the humanized or chimeric 11-1 F4 antibody comprises a constant region is derived from a human lgG1.
  • the present disclosure provides an in vivo method of detecting the presence, location, and/or amount of amyloid deposits in a patient suspected of having amyloid deposits which comprises administering to the patient an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51 ; and detecting the presence, location, and/or quantity of amyloid deposits by detection of the detectable label bound to the amyloid deposits by diagnostic imaging.
  • VH variable heavy chain
  • CDR complementarity determining region
  • VK variable
  • the disclosure provides an in vivo method of detecting the presence, location, and/or quantity of amyloid deposits in a patient suspected of having amyloid deposits which comprises administering to the patient an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48; and detecting the presence, location, and/or quantity of amyloid deposits bound to the amyloid deposits by detection of the detectable label by diagnostic imaging.
  • the disclosure provides the above-described methods wherein the method of detection is positron emission spectroscopy (PET).
  • PET positron emission spectroscopy
  • the detectable label may be 124 l or 89 Zr.
  • the antibody is selected from a chimeric or humanized 11 - 1 F4, antigen-binding fragments thereof, and the antibody CAEL-101.
  • the amyloid deposits are in the heart.
  • the present disclosure provides a composition for detecting the presence of amyloid deposits in a subject which comprises an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51.
  • VH variable heavy chain
  • CDR complementarity determining region
  • VK variable light chain
  • the present disclosure provides a composition for detecting the presence of amyloid deposits in a subject which comprises an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
  • the detectable molecule is selected from 124 l and 89 Zr.
  • the antibody is selected from chimeric or humanized 1 1 -1 F4 or antigen-binding fragments thereof and the antibody CAEL-101 .
  • the amyloid deposits are in the heart.
  • the present disclosure provides a method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprising the steps of: a) administering to said patient a composition described in the previous paragraphs and conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, b) treating the patient with a therapy intended to remove amyloid deposits, c) administering to said patient a composition described in the previous paragraphs, d) conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, and e) comparing the detected amount of detectable molecule in step d to the detected amount of detectable molecule in step a.
  • the patient may be treated in step b) with an antibody selected from a humanized or chimeric 11 -1 F4 antibody or antigen-binding fragment thereof and the antibody CAEL-101 .
  • the composition may be any of the compositions described above.
  • the method of detection may be PET.
  • the detectable label is selected from 124 l and 89 Zr.
  • the antibody is selected from humanized or chimeric 1 1 -1 F4 or antigen-binding fragments thereof and CAEL-101 .
  • the amyloid deposits are in the heart.
  • the present disclosure provides a method of determining the effectiveness of treatment to remove amyloid deposits in a patient comprising a) treating the patient with a therapeutically- effective dose of the humanized or chimeric 11-F4 antibody or antibody fragment thereof, b) administering to the patient a diagnostically-effective amount of one of the above-described compositions, and c) measuring by diagnostic imaging the amount of detectable molecule from the diagnostic composition in the lymph nodes of the patient, wherein the higher the amount of detectable molecule detected in the lymph nodes, the more effective the treatment.
  • the amyloid deposits are in the heart.
  • the chimeric 11-1 F4 antibody is CAEL-101 , the antibody produced by the CHO cells deposited with the ATCC as ACC No. PTA-125146.
  • the primary amyloidosis consists of lambda light chain fibril aggregate deposits, while in some embodiments, the primary amyloidosis consists of kappa light chain fibril aggregate deposits, and in still other embodiments, the primary amyloidosis consists of kappa and lambda light chain fibril aggregate deposits.
  • Figure 1 outlines the strategy used to clone the murine VH and VK genes from a hybridoma cell line.
  • Figure 2 is a listing of DNA and amino acid sequences of the murine 11-1 F4 antibody VH region gene, SEQ ID NO: 39 and NO: 35, respectively.
  • Figure 3 is a listing of DNA and amino acid sequences of the murine 11-1 F4 antibody VK region gene, SEQ ID NO: 40 and NO: 36, respectively.
  • Figure 4 is a map of the immunoglobulin kappa light chain expression vector pKN100. It consists of a pSV2 vector fragment, which has the SV40 early and crippled SV40 late promoter, the SV40 origin and the Co1 E1 origin. It also has the ampicillin resistance and neo genes. The crippled SV40 late promoter drives the neo genes.
  • HCMVi promoter a multiple cloning site (containing the BamH ⁇ and Hind III restriction sites) for the insertion of an immunoglobulin variable region gene, and cDNA for the human kappa constant region gene terminated by a spaC2 termination signal sequence ("Arnie"), which is in the same orientation as the kappa light chain expression cassette.
  • Figure 5 is a map of the immunoglobulin gamma 1 heavy chain expression vector pG1 D200. It consists of a pSV2dhfr vector fragment, which has the SV40 early and crippled SV40 late promoter, the SV40 origin, and the Co1 E1 origin. It also has the ampicillin resistance and dhfr genes. The crippled SV40 late promoter drives the dhfr gene. Consequently, expression is poor, allowing for the selection of multigene/high expression level clones using comparatively low levels of methotrexate. It also has the HCMVi promoter fragment, a multiple cloning site, cDNA for a human gamma 1 constant region gene (intron minus) which is followed by a spaC2 termination signal sequence ("Amie").
  • Figure 6 is a listing of the DNA and amino acid sequences of the modified murine 11-1 F4 antibody VK region gene (SEQ ID NO: 42 and NO: 47, respectively) and the sequences of the oligonucleotide primers used to modify the VK gene (SEQ ID NO: 41 and NO: 43, respectively).
  • Figure 7 is a listing of the DNA and amino acid sequences of the modified murine 11-1 F4 antibody VH region gene (SEQ ID NO: 45 and NO: 48, respectively) and the sequences of the oligonucleotide primers used to modify the VH gene (SEQ ID NO: 44 and NO: 46, respectively).
  • Figure 8 is a graphical representation of the result of the amyloid fibril binding ELISA assay.
  • the cos cell supernatants containing chimeric 11 - 1 F4 antibody were tested separately on the same ELISA plate along with purified murine 11-1 F4 antibody. The absorbance was read at OD405.
  • New sv pG1 KD200-11-1 F4.
  • New co-transfection 11-1 F4VHpG1 D200 plus 11- 1 F4VK.pKN100.
  • Figure 9 is a PET image of a mouse implanted with human cardiac derived amyloidoma which was injected with [ 124 I]CAEL-101 and imaged at day 4 after injection.
  • compositions comprising humanized antibodies, chimeric antibodies (e.g ., mouse-human antibodies) or antigen-binding fragments thereof to which in each case is linked a detectable molecule are provided that are useful for detecting the presence, location, and amount of amyloid deposits in vivo.
  • chimeric antibodies e.g ., mouse-human antibodies
  • antigen-binding fragments thereof to which in each case is linked a detectable molecule are provided that are useful for detecting the presence, location, and amount of amyloid deposits in vivo.
  • HAMA human anti-mouse antibody
  • compositions comprising at least one of said antibodies or antibody fragments linked to a detectable marker and a pharmaceutically acceptable carrier and methods of detecting the location and amount of amyloid deposits by administering to a patent an amount of said marker-linked antibody or antibody fragment effective to allow detection of amyloid deposits if such deposits are present and detecting the presence, amount, and/or location of the detectable marker bound to the amyloid deposits by diagnostic imaging.
  • the method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprises the steps of:
  • step d comparing the detected amount of detectable molecule in step c to the detected amount of detectable molecule in step a.
  • the present disclosure is directed towards measuring the extent of change in the amount of amyloid deposits of a patient (if any) by administering to said patient the marker-linked antibody or antibody fragment as described herein both prior to and after a treatment intended to remove amyloid deposits and measuring the difference in the size and extent of the detected amyloid deposits after the treatment compared to the size and extent measured prior to the treatment.
  • an in vivo method of detecting amyloid deposits in a subject comprising: a) administering to the subject a composition comprising one or more than one antibody or antigen-binding fragment thereof as described herein linked to a detectable molecule; and b) detecting by diagnostic imaging the detectable agent linked to the antibody or antigen-binding fragment thereof bound to the amyloid deposits.
  • the step of detecting may be performed using PET (positron emission tomography), SPECT (single- photon emission computed tomography), MRI, fluorescence imaging, or any other suitable diagnostic imaging method.
  • PET is the preferred imaging method.
  • This preparation of diagnostic imaging agents and their use in PET are described, for example, in Bailly, et al. - Int. J. Mol. Sci. 2017, 18, 57 ; Boerman, et al. -J Nucl Med, 2011; 52:1171-1172] and Mayer, et al. - J Nucl Med 2017; 58:538-546, all of which are incorporated herein by reference in their entirety.
  • these references are directed mainly at cancerous tumor detection, the materials and methods disclosed therein are equally useful for detecting amyloid deposits.
  • the in vivo imaging step in the methods described above may be whole body imaging for diagnostic purposes or local imaging at specific sites, such as but not limited to heart, kidney, liver, spleen, nervous system, or digestive system in a quantitative manner to assess the progression of disease or patient response to a treatment regimen.
  • the detection step in the methods as described above may be any diagnostic imaging technology including, but not limited to: positron emission tomography (PET), wherein the detectable agent is an isotope such as 11 C, 13 N, 15 0, 18 F, 64 Cu, 62 Cu, 124 l, 76 Br, 82 Rb, 89 Zr, or 68 Ga; single photon emission computed tomography (SPECT), wherein the detectable agent is a radiotracer such as 99m Tc, 111 1n, 123 l, 201 TI, or 133 Xe, depending on the specific application; magnetic resonance imaging (MRI), wherein the detectable agent may be, for example and not limited to, gadolinium, iron oxide nanoparticles and carbon-coated iron-cobalt nanoparticles.
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • MRI magnetic resonance imaging
  • antibodies are labeled with 124 l or 89 Zr.
  • the detectable molecule (sometimes described herein as a “label”, “marker”, or “agent”) as described herein may be linked, also sometimes referred to herein as "conjugated", to the antibody or fragment thereof by any suitable method known in the art.
  • the detectable agent may be linked to the antibody or antibody fragment by a covalent bond or ionic interaction.
  • the linkage may be achieved through a chemical cross-linking reaction, or through fusion using recombinant DNA methodology combined with any peptide expression system, such as bacteria, yeast or mammalian cell-based systems.
  • a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy based on the affinity of the diagnostic compositions for the amyloid deposits as determined by the uptake of the diagnostic composition by the amyloid deposits of the patient. That is, one may determine the dosing of a patient based on the detected uptake of the labeled antibody or antibody fragment. A patient showing higher uptake of the labeled antibody or antibody fragment may require a smaller amount of therapeutic antibody or antibody fragment than a patient showing a lower uptake of the labeled antibody or antibody fragment.
  • the present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the uptake of the labeled antibody or antibody fragment by the amyloid deposits, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment based on the detected amount of uptake.
  • a method of determining the effectiveness of a treatment with the chimeric or humanized antibody or antibody fragment disclosed herein to remove amyloid deposits comprises: a) treating the patient with a therapeutically-effective dose of a therapy intended to remove amyloid deposits, such as the chimeric or humanized antibody or antibody fragment disclosed herein, b) administering to the patient a diagnostically-effective amount of the diagnostic composition disclosed herein, and c) measuring the amount of detectable molecule from the diagnostic composition in the lymph nodes of the patient, wherein the higher the amount of detectable molecule detected in the lymph nodes, the more effective the treatment.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods shall mean excluding other elements of any essential significance to the composition or method.
  • Consisting of shall mean excluding more than trace elements of other ingredients for claimed compositions and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. Accordingly, it is intended that the methods and compositions can include additional steps and components (comprising) or alternatively including steps and compositions of no significance (consisting essentially of) or alternatively, intending only the stated method steps or compositions (consisting of).
  • “about” means plus or minus 10%.
  • the terms“individual”,“patient”, or“subject” can be an individual organism, a vertebrate, a mammal (e.g ., a bovine, a canine, a feline, or an equine), or a human.
  • the individual, patient, or subject is a human.
  • an“isolated antibody” is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities ⁇ e.g., an isolated antibody that specifically binds to an amyloid fibril is substantially free of antibodies that do not bind to amyloid fibrils).
  • An isolated antibody that specifically binds to an epitope of an amyloid light chain fibril ⁇ e.g., a kappa and/or lambda fibril) may, however, have cross- reactivity to other proteins, such as amyloid A fibrils. However, the antibody preferably always binds to human amyloid light chain fibrils.
  • an isolated antibody is typically substantially free of other cellular material and/or chemicals.
  • the phrase “diagnostically effective amount” means the amount of detectable marker-linked antibody or antibody fragment which, when administered to a patient or subject, permits in vivo detection, localization, and/or quantification by diagnostic imaging of amyloid deposits in the patient or subject if any such deposits are present. It is emphasized that a diagnostically effective amount will not always be effective to detect amyloid deposits, even though such amount is deemed to be a diagnostically effective amount by those of skill in the art. The diagnostically effective amount may vary based on the route of administration and dosage form, the age and weight of the subject, and/or the subject’s condition, including the type and stage of the amyloidosis at the time that diagnosis commences, among other factors.
  • humanized antibody refers to an antibody that comprises the CDRs of antibodies derived from mammals other than human, and the framework region (FR) and the constant region of a human antibody.
  • a humanized antibody is useful as a component in a diagnostic composition according to the present disclosure since antigenicity of the humanized antibody in human body is lowered.
  • the term“antibody fragment” refers to a part of an antibody that comprises a CDR of the antibody but has been engineered to delete some of the structure of the intact antibody.
  • Engineering antibody fragments is well known in the art, as shown (for example) in Holliger, et al. - Nature Biotechnology: 23 (9), 1126-1135 (2005), incorporated herein by reference in its entirety.
  • the term“antigen-binding fragment” as used herein means an antibody fragment that binds to amyloid deposits.
  • the term “pharmaceutically-acceptable carrier” means a material for admixture with a pharmaceutical or diagnostic compound (e.g., a chimeric antibody linked to a detectable molecule) for administration to a patient as described, for example, in“Ansel’s Pharmaceutical Dosage Forms and Delivery Systems”, Tenth Edition (2014).
  • detecttable marker refers to substances that may be detected in vivo by diagnostic imaging techniques, such as (but not limited to) PET, SPECT, and MRI.
  • the terms“diagnostic imaging” or“imaging” mean methods of in vivo imaging of a detectable molecule in a patient or subject to determine the presence, location and/or amount of a tissue or substance (such as an amyloid deposit) in the patient or subject to which the detectable molecule has been localized, such as by being linked to an antibody or antibody fragment that binds to the tissue or substance to be detected.
  • a tissue or substance such as an amyloid deposit
  • Murine antibodies are generally unsuitable for administration to other animal species (such humans) because the receiving species will recognize the murine antibody as antigenic and will produce antibodies against it.
  • the antigenicity of an antibody from one species when injected into another species is normally caused by a portion of a constant domain. Such an antigenic response will impede or prevent the desired therapeutic or diagnostic effect of the murine antibody.
  • this antigenic response is called human anti-mouse antibody (HAMA).
  • HAMA human anti-mouse antibody
  • the antibodies described in the‘594 patent have the potential to be highly immunogenic in humans via the human anti-mouse antibody (HAMA) response. Since the HAMA response usually results in the rapid clearance of a mouse antibody from the human recipient, HAMA would severely limit any potential human therapeutic or diagnostic benefit a murine antibody could have.
  • the present disclosure provides compositions and methods for detecting and/or monitoring amyloid deposition diseases that is less likely to produce an immunogenic HAMA response in a patient following administration.
  • compositions comprising humanized and chimeric antibodies or antigen-binding fragments thereof having linked thereto a detectable molecule.
  • the disclosed compositions are useful for imaging the location and amount of amyloid deposits in a subject.
  • an antibody consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two copies of a light (L) chain polypeptide.
  • each heavy chain contains one N-terminal variable (VH) region and three C-terminal constant (CH1 , CH2 and CH3) regions
  • each light chain contains one N-terminal variable (VL or VK) region and one C-terminal constant (CL) region.
  • Each variable domain of the light and heavy chain in an antibody also comprises three segments called complementarity-determining regions (“CDR”) or hypervariable regions.
  • CDR complementarity-determining regions
  • the variable regions of each pair of light and heavy chains form the antigen binding site of an antibody, whereas the constant region provides structural support and modulates the immune response initiated by the antigen binding.
  • Chimeric antibodies incorporate the variable region of a non-human antibody into the constant region of a human antibody.
  • a chimeric 11-1 F4 antibody may be created by expressing the murine variable region with the Fc region of a human antibody, such as a human lgG1.
  • Humanized forms of non-human (e.g ., murine) antibodies can be obtained, which contain minimal sequences derived from non-human immunoglobulin.
  • a humanized antibody may comprise one or two or more variable domains in which variable regions are derived from non-human immunoglobulin and framework regions (FR) correspond to a human immunoglobulin sequence.
  • FR framework regions
  • a humanized anti-AL antibody comprises a human antibody framework region.
  • Such antibodies can be prepared by known techniques.
  • the murine 11-1 F4 monoclonal antibody is an anti-AL antibody produced by the SP2/0 hybridoma cell deposited by Alan Solomon, MD (University of Tennessee Medical Center at Knoxville, TN).
  • the hybridoma cell line is available from the American Type Culture Collection (ATCC access PTA- 105).
  • the V K region (SEQ ID NO: 36) and the V H region (SEQ ID NO: 35) of the 11-1 F4 antibody are shown in Table 1 below.
  • the CDR sequences for the heavy and light chains and provided in Table 2.
  • the chimeric 11-1 F4 antibody binds to an epitope expressed by the b-pleated sheet configuration of amyloids, just as its murine counterpart does, but surprisingly, as shown in Example 6 below, the chimeric antibody binds to AL amyloid fibrils with higher affinity than the 11-1 F4 mouse antibody from which it was derived.
  • the humanized form may also have a binding affinity for amyloid fibrils that is higher than that of the murine counterpart.
  • the disclosed humanized and chimeric antibodies may utilize all different types of human constant regions and/or framework regions.
  • the disclosed humanized and chimeric antibodies may comprise the constant regions and/or framework regions of a human IgG (including lgG1 , lgG2, lgG3, and lgG4), IgA, IgE, IgH, or IgM.
  • the disclosed humanized or chimeric 11-1 F4 antibody comprises a human lgG1 constant region.
  • the disclosed antibodies may comprise one or more substitutions, insertions, or deletions, so long as the antibody maintains the ability to bind to amyloid fibrils (e.g ., kappa and/or lambda light chain fibrils).
  • amyloid fibrils e.g ., kappa and/or lambda light chain fibrils.
  • a chimeric 11 -1 F4 antibody of the present disclose may comprise heavy and light chains with about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91 %, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% identity compared to the corresponding heavy and light chain sequences disclosed herein, so long as the antibody maintains the ability to bind to amyloid fibrils.
  • a humanized 11 -1 F4 antibody of the present disclose may comprise CDRs that have about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91 %, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% identity compared to the corresponding CDR sequences disclosed herein, so long as the antibody maintains the ability to bind to amyloid fibrils.
  • DMEM Dulbecco's Modified Eagles Medium
  • FBS Fetal Bovine Serum
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • DNA deoxyribonucleic acid
  • cDNA copy DNA
  • PCR polymerase chain reaction
  • min minute
  • sec second
  • Amino acids are represented by the IUPAC abbreviations, as follows: Alanine (Ala), Arginine (Arg), Asparagine (Asn), Aspartic acid (Asp), Cysteine (Cys), Glutamine (Gin), Glutamic acid (Glu), Glycine (Gly), Histidine (His), Isoleucine (lie), Leucine (Leu), Lysine (Lys), Methionine (Met), Phenylalanine (Phe), Proline (Pro), Serine (Ser), Threonine (Thr), Tryptophan (Trp), Tyrosine (Tyr), Valine (Val).
  • nucleotides Adenine (A), Cytosine (C), Guanine (G), Thymine (T), Uracil (U), Adenine or Guanine (R), Cytosine or Thymine (Y), Guanine or Cytosine (S), Adenine or Thymine (W), Guanine or Thymine (K), Adenine or Cytosine (M), Cytosine or Guanine or Thymine (B), Adenine or Guanine or Thymine (D), Adenine or Cytosine or Thymine (H), Adenine or Cytosine or Guanine (V), and any base (N).
  • the murine 11-1 F4 monoclonal antibody heavy and kappa light chain variable region genes described in United States patent 8,105,594 were PCR modified to facilitate the expression of the chimeric 11-1 F4 antibody in mammalian cells.
  • modified variable region genes were cloned into the appropriate mammalian expression vectors, creating the constructs 11 - 1 F4VHpG1 D200 and 11 -1 F4VK.pKN100.
  • a single supervector construct, pG1 KD200-11-1 F4 was made from the 11-1 F4VHpG1 D200 and 11 - IF4VK.pKN100 constructs by EcoRI restriction enzyme digest and ligation.
  • the chimeric 11 -1 F4 antibody was transiently expressed in COS cells by both cotransfection and single supervector transfection.
  • an antibody consists of four polypeptides: two identical copies of a heavy (FI) chain polypeptide and two copies of a light (L) chain polypeptide.
  • each heavy chain contains one N-terminal variable (VH) region and three C-terminal constant (CH1 , CH2 and CH3) regions
  • each light chain contains one N-terminal variable (VL or VK) region and one C- terminal constant (CL) region.
  • the variable regions of each pair of light and heavy chains form the antigen binding site of an antibody.
  • An antibody useful in the compositions and methods of the invention may be a chimeric mouse-human monoclonal antibody comprising the VK region of SEQ ID NO: 47 and the VH region of SEQ ID NO: 48 or a humanized monoclonal antibody comprising CDR sequences of SEQ ID NOs: 49-54.
  • These antibodies bind to an epitope expressed by the b-pleated sheet configuration of amyloid fibrils.
  • the antibodies bind to this epitope with higher affinity than the 11-1 F4 mouse antibody from which they were derived, which comprises the VK region of SEQ ID NO: 36 and the VH region of SEQ ID NO: 35.
  • the invention includes methods of detecting amyloid deposits in a human patient which comprises administering to the patient a diagnostically effective dose of one of the above antibodies linked to a detectable molecule in a pharmaceutically-acceptable carrier.
  • the antibody composition may be administered by any conventional route of administration, but parenteral administration (such as intravenous) is preferred.
  • Pharmaceutically-acceptable carriers are well-known in the art and a suitable one can be selected by one of skill in the medical field.
  • Materials useful to make the subject antibody include vector constructs selected from the group consisting of 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200, shown in Figures 5 and 6, respectively, and the superconstruct pG.1 KD20011-1 F4 made from the two above vector constructs.
  • Other useful materials include the modified murine 11-1 F4 antibody VK region gene (SEQ ID NO: 42) and the modified 11- 1 F4 antibody VH region gene (SEQ ID NO: 45), as well as the respective primers SEQ ID NO: 41 , 43, 44, and 46.
  • the subject antibody may be made by co-transfection of the vector constructs 11 -1 F4VK.pKN100 and 11 - F4VFI.pG1 D200 or the superconstruct pG.1 KD20011 -1 F4 in a suitable mammalian host cell, such as COS (Chinese hamster ovary) cells.
  • a suitable mammalian host cell such as COS (Chinese hamster ovary) cells.
  • Diagnostic compositions suitable for use in the methods described herein can include the disclosed humanized or chimeric 11-1 F4 antibodies, humanized antibodies, or antigen-binding antibody fragments linked in each case to a detectable marker, and a pharmaceutically acceptable carrier or diluent.
  • the composition may be formulated for intravenous, subcutaneous, intraperitoneal, or intramuscular administration, although intravenous administration is preferred.
  • Pharmacologically acceptable carriers for various dosage forms are known in the art.
  • solvents, solubilizing agents, suspending agents, isotonicity agents, buffers, and soothing agents for liquid preparations are known.
  • the pharmaceutical compositions include one or more additional components, such as one or more preservatives, antioxidants, stabilizing agents and the like.
  • Sterile injectable solutions can be prepared by incorporating the marker-linked antibody or antibody fragment in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration.
  • dispersions are prepared by incorporating the diagnostic composition into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • amyloidosis is caused by the buildup of an abnormal protein called amyloid.
  • Amyloid is produced in the bone marrow and can be deposited in any tissue or organ. The specific cause of the condition depends on the type of amyloidosis.
  • amyloidosis or amyloid diseases There are several types of amyloidosis or amyloid diseases, including AL amyloidosis, AA amyloidosis, and hereditary amyloidosis.
  • AL amyloidosis is the most common type and can affect the heart, kidneys, skin, nerves and liver. Previously known as primary amyloidosis, AL amyloidosis occurs when the bone marrow produces abnormal antibodies that cannot be broken down. The antibodies are deposited in various tissues as amyloid plaques (amyloid deposits), which interfere with normal function of the tissue or organ.
  • AA amyloidosis generally affects the kidneys but occasionally also affects the digestive tract, liver or heart. It was previously known as secondary amyloidosis. It often occurs along with chronic infectious or inflammatory diseases, such as rheumatoid arthritis or inflammatory bowel disease.
  • Hereditary amyloidosis is an inherited disorder that usually often affects the liver, nerves, heart, and/or kidneys. Many different types of gene abnormalities present at birth are associated with an increased risk of amyloid disease or hereditary amyloidosis. The type and location of an amyloid gene abnormality can affect the risk of certain complications, the age at which symptoms first appear, and the way the disease progresses over time.
  • Amyloid deposits or plaques reduce the heart’s ability to fill with blood between heartbeats. Less blood is pumped with each beat, and this may lead to shortness of breath. Amyloid deposits or plaques in or around the heart may also cause irregular heartbeats and congestive heart failure, among other organ dysfunctions
  • kidneys When an amyloid disease affects the kidneys, it will often harm the kidneys’ filtration ability, allowing protein to leak from the blood into the urine (i.e., proteinuria). Moreover, the kidneys’ ability to remove waste products from your body is lowered, which may eventually lead to kidney failure.
  • amyloid deposition disease such as primary (AL) amyloidosis
  • a patient e.g ., a human patient
  • a humanized or chimeric 11-1 F4 antibody or an antigen-binding fragment thereof linked to a detectable marker together with a pharmaceutically acceptable carrier, in an amount effective to detect, locate, and/or quantify amyloid deposits if such are present, followed by detecting the detectable molecule bound to the amyloid deposits in the patient by diagnostic imaging to determine the extent, location, and/or amount of the amyloid deposits if such are present.
  • Also provided herein is a method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprising the steps of:
  • a therapy intended to remove amyloid deposits such as a humanized or chimeric 11 -1 F4 antibody or antigen-binding fragment thereof
  • step d comparing the detected amount of detectable molecule in step c to the detected amount of detectable molecule in step a.
  • the above monitoring method can be conducted starting before any treatment has taken place or can be used in the midst of a treatment regimen. That is, step a) above can be performed prior to any treatment of the patient to determine a baseline amount of amyloid deposit or step a) above can be conducted after one or more courses of treatment to determine the effect of the next succeeding course of treatment.
  • the amyloid deposition disease e.g ., primary amyloidosis
  • the disclosed methods comprise monitoring the treatment of a patient suffering from relapse or refractory ALA.
  • the patient may have kappa ALA.
  • the patient may have lambda ALA.
  • Exemplary diagnostic amounts can vary according to the size and health of the individual being treated, as well as the condition being diagnosed.
  • the diagnostically effective amount of a radiolabeled humanized or chimeric 11 -1 F4 antibody for PET is about 1-10 mCi.
  • the dose may be higher or lower.
  • One of skill in the diagnostic imaging art would know how to select an appropriate amount of labeled antibody or antibody fragment, as described (for example) in the references listed above.
  • the murine 11-1 F4 monoclonal antibody heavy and light chain variable region genes were PCR cloned and a detailed sequence analysis of all variable region genes isolated (both pseudo and functional) was performed. Detailed DNA and amino acid sequences of the murine 11-1 F4 antibody heavy and light chain variable region genes were obtained. Materials
  • RNA solution kit was obtained from Stratagene (USA), while the first strand cDNA synthesis kit was purchased from Pharmacia (UK). All the constituents and equipment for the RCR-reaction, including AmpliTaq® DNA polymerase, were purchased from Perkin Elmer (USA).
  • the TOPO TA Cloning ® kit was obtained from Invitrogen (USA). Agarose (UltraPureTM) was obtained from Life Technologies (UK).
  • the ABI PRISM® Big DyeTM terminator cycle sequencing ready reaction kit pre-mixed cycle sequencing kit and the ABI PRISM® 310 sequencing machine were both purchased from PE Applied Biosystems (USA). All other molecular biological products were obtained from New England Biolabs (USA) and Promega (USA).
  • B2C4 and B2D6 Two clones (B2C4 and B2D6) of the SP2/0 hybridoma cell line producing the ct-human light chain monoclonal antibody 11 -1 F4, were kindly provided by Alan Solomon, MD (University of Tennessee Medical Center at Knoxville, TN).
  • the hybridoma cell line is available from the American Type Culture Collection (ATCC access PTA-105).
  • the cell lines were cultured using DMEM media supplemented with 20% (v/v) FBS, penicillin/streptomycin and L- Glutamine. Cells were cultured until a total viable cell count of 10 8 cells was reached.
  • the cells were harvested separately from each clone as follows.
  • the mouse hybridoma cell line was grown in suspension in an appropriate culture medium and in sufficient quantities to provide a total viable cell count of about 10 8 cells.
  • the culture supernatant was harvested and the hybridoma cells pelleted in a bench top centrifuge (250 g, 5 min). The cells were gently re-suspended in 20 ml PBS and a 100 pi aliquot was taken for a viable cell count.
  • the cells in the aliquot were pelleted once more and 200 mI of PBS and 200 mI of trypan blue were added to the 100 mI of cells and mixed gently.
  • RNA was then isolated separately from the cells of each clone using a Stratagene RNA isolation kit, according to the manufacturer’s instructions.
  • One ml of 2 M sodium acetate (pH 4.0) was added to the sample and the contents of the tube were thoroughly mixed by repeatedly inverting the tube.
  • To the tube was added 10.0 ml of phenol (pH 5.3-5.7) and the contents again mixed thoroughly by inversion.
  • To the mixture was added 2.0 ml of chloroform-isoamyl alcohol mixture, the tube was capped and vigorously shaken for 10 seconds, and the tube was incubated in ice for 15 minutes.
  • the sample was transferred to a 50-ml thick-walled, round-bottom centrifuge tube that had been pre-chilled on ice and the tube was spun in a centrifuge at 10,000 x g for 20 minutes at 4°C. Two phases were visible in the tube after centrifugation.
  • the upper, aqueous phase contained the RNA, while the lower phenol phase and interphase contained DNA and proteins.
  • the RNA- containing upper, aqueous phase was transferred to a fresh centrifuge tube and the lower phenol phase was discarded.
  • An equal volume of isopropanol was added to the aqueous phase and the contents mixed by inversion, following which the tube was incubated for 1 hour at -20°C to precipitate the RNA.
  • the tube was spun in a centrifuge at 10,000 x g for 20 minutes at 4°C. After centrifugation, the pellet at the bottom of the tube, which contains the RNA, was removed and the supernatant discarded. The pellet was dissolved in 3.0 ml of solution D, 3.0 ml of isopropanol was added to the tube and the contents mixed well. After incubating the tube for 1 hour at -20°C, it was again spun in a centrifuge at 10,000 x g for 10 minutes at 4°C and the supernatant removed from the tube and discarded.
  • RNA had been protected from ribonucleases by the presence of guanidine isothiocyanate but was now no longer protected.
  • the pellet was washed with 75% (v/v) ethanol (DEPC-treated water (25%)) and the pellet was dried under vacuum for 2-3 minutes. The RNA pellet is re-suspended in 0.5-2 ml of DEPC-treated water.
  • an Amersham Pharmacia Biotech first strand cDNA synthesis kit was employed to produce a single- stranded DNA copy of the 11-1 F4 hybridoma mRNA using the Not I- d(T) 18 primer supplied with the kit.
  • One reaction was performed for each of the two RNA samples isolated, as follows.
  • the components used were: Bulk first strand cDNA reaction mix, Cloned FPLCpureTM Murine Reverse Transcriptase, RNAguardTM, BSA, dATP, dCTP, dGTP, and dTTP, 200 mM DIT aqueous solution, Not l-d(T) 18 primer: 5' d[AACTGGAAGAATTCGCGGCCGCAGGAAi 8 ]-3', and DEPC treated water.
  • RNA in 20 pi DEPC water was heated to 65 'C for I0 min and then chilled on ice.
  • the bulk first strand cDNA reaction mix was pipetted gently to obtain a uniform suspension and the reaction set up in a 0.5 ml microcentrifuge tube as below.
  • the reactants were mixed gently by pipetting and incubated 37°C for 1 hour.
  • VH genes and VK genes were then PCR amplified from the ssDNA template using the method described by Jones and Bendig (Bio/Technology 9:88).
  • each tube a 100 pi reaction mixture was prepared, each reaction mixture containing 69.3 mI of sterile water, 10 mI of 10 X PCR buffer II, 6 mI of 25 mM MgC , 2 mI each of the 10 mM stock solutions of dNTPs, 2.5 mI of 10 mM MKC primer, 2.5 mI of one of the 10 mM MKV primers and 1 mI of RNA-cDNA template mix. To each of the tubes was then added 0.7 mI of AmpliTaq® DNA polymerase and the completed reaction mix overlaid with 50 mI of mineral oil.
  • a similar series of reaction mixes was prepared as described above to PCR-clone the mouse heavy chain variable region gene. However, this time twelve reaction tubes were labeled and one of the twelve MHV primers and the appropriate MHC primer were added to each. That is, to PCR-amplify the variable domain gene of a mouse y1 heavy chain, for example, the MHC G1 primer was used.
  • reaction tubes were loaded into a DNA thermal cycler and cycled (after an initial melt at 94 °C for 1.5 min) at 94 °C for 1 min, 50 °C for I min and 72 °C for 1 min over 25 cycles. The last cycle was followed by a final extension step at 72 °C for 10 min before cooling to 4 °C. Except for between the annealing (50 °C) and extension (72 °C) steps when an extended ramp time of 2.5 min was used, a 30 sec ramp time was used between each step of the cycle.
  • Table 1 - PCR primers for cloning mouse kappa light chain variable region genes
  • Those putative positive clones so identified were double-stranded plasmid DNA sequenced using the ABI PRISM 310 Genetic Analyzer and the ABI PRISM BigDyeTM terminator.
  • Three positive clones each of the VH and VK genes from the B2C4 hybridoma cell line clone were sequenced, as were four positive clones of the V K gene and six of the VH gene from the B2D6 hybridoma cell line clone.
  • the degenerate leader sequence primer MHV7 in combination with a mix of the MHCGI-3 constant region primers (Table 1 ), yielded a PCR product of about 600 bp from template cDNAs derived from both the B2C4 and B2D6 hybridoma cell lines. Since this band was larger than the expected size for an average V hi gene (450 bp), it was not investigated further.
  • the degenerate leader sequence primer MHV6 in combination with a mix of the MHCGI-3 constant region primers (Table 1 ), yielded a PCR product of the expected size (450 bp) for a VH gene from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
  • Table 4 shows the results of the PCR amplifications performed to clone the murine 11-1 F4 monoclonal antibody variable region heavy (a) and light (b) chain genes from the SP2/0 hybridoma cell lines B2C4 and B2D6. Column three contains a record of the actual PCR results. Where a band was observed for a particular combination of primers its size in base pairs (bp) was recorded in the appropriate space.
  • the degenerate leader sequence primer MKV2 in combination with the MKC constant region primer (Table 2), produced a PCR product which was smaller than the expected 450 bp band (when viewed on an agarose gel) from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
  • the MKV2 primer amplified a well known kappa light chain pseudogene. Therefore, sequence analysis of one clone of each PCR product was performed in order to confirm that this product was a pseudogene and not the murine 11-1 F4 antibody VK gene. This sequence analysis revealed that this PCR clone was indeed the pseudogene.
  • the degenerate leader sequence primer MKV1 in combination with the MKC constant region primer (Table 1 ), produced a PCR product of about the expected size (450 bp) for a VK gene, from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
  • the 11-1 F4 antibody heavy chain variable region gene was cloned (using constant region specific and leader sequence specific primers) from the hybridoma mRNA and sequenced.
  • the murine 11-1 F4 antibody variable region kappa light chain gene was also successfully cloned and sequenced, as was a non-functional VK pseudogene gene.
  • This pseudogene was first identified by Carroll et al (Molecular Immunology (1988) 25:991 ). The sequence arises from an aberrent mRNA transcript which is present in all standard fusion partners derived from the original MOPC-21 tumor (including SP2/0). As a result of the aberrant mRNA, the invariant cysteine at position 23 is replaced by a tyrosine residue, and the VJ joint is out of frame, resulting in a stop codon at position 105.
  • the back (BAK) primers F39836 and F39835 introduced a Hind ⁇ restriction site, a Kozak translation initiation site, and an immunoglobulin leader sequence to the 5' ends of the VK and VH genes respectively.
  • the forward (FOR) oligonucleotide primer F39837 introduced a splice donor site and a BamH ⁇ restriction site to the 3' end of the VK gene while the forward (FOR) oligonucleotide primer F58933 appended the first 22 base pairs of the gamma-1CHi gene including an Apa ⁇ restriction site to the 3' end of the VH gene.
  • Table 5 - Oligonucleotide primers used to PCR modify the 11 -1 F4 heavy and kappa light chain variable region genes
  • the Kozak consensus sequence is crucial to the efficient translation of a variable region sequence (Kozak - J Mol Bio 196:947). It defines the correct AUG codon from which a ribosome commences translation, and the single most critical base is the adenine (or less preferably, a guanine) at position -3, upstream of the AUG start.
  • the immunoglobulin leader sequence ensures that the expressed antibody is secreted into the medium and therefore is easily harvested and purified.
  • the leader sequences used in this instance were the murine 1 1 -1 F4 ⁇ A ⁇ and VH leader sequences cloned from the hybridoma cDNA during the VH and VK cloning process.
  • the splice donor sequence is important for the correct in-frame attachment of the light chain variable region to its appropriate constant region, thus splicing out the 130 bp VK:CK intron.
  • the heavy chain variable region was attached directly to its appropriate constant region gene via the Apa ⁇ site, thus eliminating the need for a splice donor site.
  • PCR reactions were prepared as follows, one for each variable region gene.
  • the plasmids 11-1 F4 VH.PCR2.1 and 11 -1 F4 VK.PCR2.1 described above were used as templates.
  • a 100 pi reaction mixture was prepared in each PCR tube, each mixture containing up to 41 pi of sterile water, 10 mI of 10 x PCR buffer I, 8 mI of the 10 mM stock solution of dNTPs, 1 m I of 10 mM of 5' forward primer, 1 mI of the 10 mM 3' Reverse primer, and 1 m I of a 1/10 dilution of template DNA.
  • Colonies containing the plasmid with a correctly sized insert were identified by PCR-screening the colonies using the 1212 and 1233 oligonucleotide primers (Table 3) according to the method of Gilssow and Clackson. Those putative positive clones so identified were double-stranded plasmid DNA sequenced using the ABI PRISM 310 Genetic Analyzer and the ABI PRISM BigDyeTM terminator. Two positive clones each of the Topo TA cloned V hi and V K genes were sequenced.
  • the modified 11-1 F4 VH gene was also subcloned as a Hind ⁇ -Apa ⁇ fragment into the expression vector pG1 D200 (Figure 5); this vector contained a human y1 constant region gene (allotype: G1 m (-1 Glu377, Met38l, -2 Ala462, 3 Arg222, Ser229)). Both the kappa and y1 constant region allotypes used are commonly found in the Caucasian population.
  • the ligated expression constructs, 11-1 F4VK.pKN100 and 11- 1 F4VH.pG1 D200, were then used to transform DH5ct competent cells, and positive clones were identified using the PCR screening method discussed above with the original PCR modification primers (Table 4). The expression vectors are readily available.
  • a single supervector expressing both immunoglobulin chains of the chimeric 11-1 F4 antibody was constructed as follows.
  • the 11-1 F4 kappa light chain expression cassette (which contained the HCMVi promoter, the 11- 1 F4 kappa light chain variable region gene, and the kappa light chain constant region gene) was restriction enzyme digested (EcoRI at positions 1 and 2490) out of the 11-1 F4VK.pKN100 construct ( Figure 4) and subsequently ligated into the 11-1 F4VHpG1 D200 construct via the unique EcoRI (position 4297, Figure 5). This ligation resulted in the construction of a supervector construct, pG1 KD200-11-1 F4, containing both the heavy and kappa light chains of the 11 -1 F4 chimeric antibody.
  • the chimeric 11-IF4 antibody was transiently expressed in COS cells from the European Collection of Cell Cultures (ECACC) in two ways:
  • the following transfection method was used.
  • the COS cell line was grown in DMEM supplemented with 10% (v/v) FCS, 580 pg/ml L- glutamine and 50 Units/ml penicillin/ 50 pg/ml streptomycin (“media”) in a 150 cm 2 flask until confluent.
  • the cells were trypsinized, spun down in a bench top centrifuge (250 g for 5 min), then re-suspended in 6 ml of media before dividing them equally between three 150 cm 2 flasks, each containing 25 ml of fresh, pre-warmed media.
  • the cells were incubated overnight at 37 °C in 5% CO2 and then harvested the next day while they are still growing exponentially.
  • Each flask contained approximately 1 x 10 7 cells.
  • the cells were trypsinized again, pelleted as before, and washed in 20 ml of PBS, following which they were re-suspend in sufficient PBS to create a cell concentration of 1 x I0 7 cells/ml.
  • 700 pi of these washed COS cells were pipetted into a Gene Pulser® cuvette, to which was then added 1 mI of both the heavy chain and kappa light chain expression vector DNA (each at 10 pg) or 13 pg of the super-vector construct.
  • a 1900 Volt, 25 pFarad capacitance pulse was delivered to the mixture using the Bio-Rad Gene Pulser® apparatus.
  • the pulsing was repeated for each experimental transfection and a "no DNA" control (in which the COS cells were electroporated in the absence of any DNA).
  • a positive control of a previously-expressed antibody was also carried out to test the efficiency of the COS cells.
  • the COS cells were allowed to recover at room temperature for 10 min, then gently pipetted the into a 10 cm diameter tissue culture dish containing 8 ml of pre- warmed DMEM supplemented with 10% (v/v) y - globulin free FBS, 580 pg/ml L- glutamine and 50 Units/ml penicillin / 50 pg/ml streptomycin, and incubated in 5% CO2 at 37 °C for 72 hours before harvesting the COS cell supernatant for analysis. After incubation for 72 hours the medium was collected, spun to remove cell debris and analyzed by ELISA for chimeric antibody production and antigen binding of the c11-1 F4 antibody.
  • IgG molecules present in the COS cell supernatant were quantified using a capture ELISA assay.
  • IgG molecules were captured on a Nunc-lmmuno MaxiSorbTM plate via an immobilized goat anti-human IgG, Fey fragment - specific antibody, and detected via an anti-human kappa light chain peroxidase conjugated antibody.
  • a standard curve was generated by capturing and detecting known concentrations of a standard IgG antibody on the same plate in the same way as follows. Each well of a 96-well immunoplate was coated with 100 mI aliquots of 0.4 pg/ml goat anti-human IgG antibody diluted in PBS and incubated overnight at 4 °C.
  • the chimeric 11-1 F4 antibody was tested for binding to amyloid fibrils using a direct binding ELISA assay.
  • Synthetic fibrils were formed from an immunoglobulin light chain protein and used to monitor the reactivity of the antibody in a solid-state ELISA-based assay using a "low- binding" polystyrene plates (Costar,# 3474).
  • coating buffer 0.1 % bovine serum albumin in phosphate buffered saline pH 7.5.
  • the sample was then sonicated for 20 sec using a Tekmar Sonic Disruptor sonicating probe, with the power set to 40% of maximum, resulting in a solution of short fibrils composed of up to 2-5 protofiliments each.
  • This solution was then diluted to 5 ml, mixed well by vortex, and aliquoted into the wells of the plate. This process yielded 50 pi of fibril solution having a concentration of 50 pg/ ml in each well.
  • the plate was then dried overnight by placing it uncovered in a 37 °C incubator.
  • the ELISA assay was then performed as follows within 48 hours of preparing the plate.
  • the wells were blocked by the addition of 100 pi of l% BSA in PBS and incubated for 1 hour at room temperature on a shaker.
  • the plate was washed x3 in PBS, 0.05% Tween 20 (v/v).
  • To each well of the plate was added 50 pi of a solution of c11-1 F4 (3pg/ml antibody in 0.1% BSA/PBS) and the plate incubated at room temperature for 1 hour on a shaker.
  • the plate was again washed x3 (as before) and detection of bound antibody was accomplished using a biotinylated goat anti-mouse IgG antibody (Sigma# B- 8774, anti-heavy and light-chain).
  • the 11-1 F4VK.pKN100 and 11-1 F4VHpG1 D200 constructs were also subsequently used to construct a single supervector (pG1 KD200-11-1 F4), which expressed the chimeric 11 - 1 F4 antibody in mammalian cells.
  • the chimeric 11-1 F4 antibody expression levels, from both cotransfections and supervector transfections of ECACC COS cells were assayed.
  • the chimeric 11-1 F4 antibody was tested for binding to target antigen (amyloid fibrils kindly supplied by the NCI) by direct binding ELISA.
  • target antigen amyloid fibrils kindly supplied by the NCI
  • the results of the binding ELISA are presented in Figure 8. Supenatants from the two best individual pG1 KD200- 11-1 F4 supervector transfections were assayed in parallel with one supernatant from the corresponding co-transfection.
  • cGMP grade CAEL-101 was radiolabeled with 124 l, a positron emitting radioisotope used for PET imaging, with the standard iodegen reaction. (Fraker, et al. - Biochem Biophys Res Commun 80(4): 849-57; Markwell et at. - Biochem 17:4807-17) Approximately 5 days after human amyloid extract was implanted to form subcutaneous amyloidomas, the mice were injected with 50-200pCi of [ 124 l ]CAEL-101 and imaged 1 , 4 and 7 days post injection using an Inveon microPET scanner. SUVmax for amyloidomas and contralateral background were obtained by drawing regions of interest in the PMOD software package and calculating tumor-to-background (amyloid deposit-to-background) (T:B) ratios at 1 and 4 days post tracer infusion.
  • T:B tumor-to-background ratios at 1 and 4 days post tracer infusion.
  • [00153] [ 124 I]CAEL-101 successfully imaged 100% of mice bearing human amyloid extracts (k1 , A1 and A6 subtypes derived from heart, liver, spleen, and kidney). We demonstrated for the first time imaging of cardiac derived amyloidosis (shown in Figure 9), which was reported to not work in prior literature. (Wall, et at. - Blood. 2010 116:2241 ). Human amyloidomas were visualized at both 1 and 4 days post tracer infusion, with significantly increasing T:B ratio by day 4. T:B ratios ranged from 2.1 to 4.2 at 4 days. We found heterogeneous uptake among various amyloidomas.
  • mice implanted with k subtypes demonstrated significantly better in vivo T:B ratios (4.1 +/- 0.20), compared to l subtypes (2.8 +/- 0.46), although all amyloidomas exhibited T:B uptake > 2.1 , which would be clinically significant.
  • This result of heterogeneous uptake among the various amyloidomas supports using real time PET imaging to stratify patients for therapy with the chimeric or humanized antibody described herein as well as to determine the appropriate dose of the antibody therapy. That is, one may determine the dosing of a patient based on the detected uptake of the labeled antibody or antibody fragment.
  • a patient showing strong uptake (affinity) for the labeled antibody or antibody fragment may require a smaller amount of therapeutic antibody or antibody fragment than a patient showing a weaker uptake (affinity) for the labeled antibody or antibody fragment.
  • the present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the uptake of the labeled antibody or antibody fragment by the amyloid deposits of the patient, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment to the patient based on the determined amount of uptake of the labeled antibody or antibody fragment.

Abstract

Methods and diagnostic compositions for detection of amyloid deposits using a chimeric (e.g., mouse-human) antibody or antigen-binding fragment thereof linked to a detectable label are disclosed.

Description

METHODS AND COMPOSITIONS FOR IMAGING AMYLOID DEPOSITS
REFERENCE TO A SEQUENCE LISTING SUBMITTED BY EFS-WEB
[0001] The contents of the ASCII text file of the sequence listing named “8441 -0018-1 -ST25”, which is 16.9 kb in size, was created on January 22, 2019, and was electronically submitted via EFS-WEB with this application, is incorporated herein by reference in its entirety.
CLAIM FOR PRIORITY
[0002] This application claims priority from United States provisional application 62/753,410, filed October 31 , 2018, which is incorporated herein in its entirety.
FIELD OF THE INVENTION
[0003] The present disclosure relates to humanized and chimeric ( e.g ., mouse-human) antibodies and antigen-binding fragments thereof having a detectable molecule linked or conjugated thereto and to methods of using the same to detect and image amyloid deposits.
BACKGROUND OF THE INVENTION
[0004] The following discussion is provided merely to aid the reader in understanding the disclosure and is not admitted to describe or constitute prior art thereto.
[0005] Native antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons composed of two identical light chains and two identical heavy chains. Each light chain is linked to a heavy chain by one disulfide bond, while the number of additional disulfide linkages between the heavy chains varies with different antibody isotypes. The simplest isotype is IgG, which comprises just two light chains and two heavy chains, in which the two heavy chains are linked by two disulfide linkages. Each heavy chain has a variable domain (VH) at one end with a number of adjacent constant domains. Each light chain has a variable domain (VL) at one end and a constant domain at its other end. Each variable domain of the light and heavy chain in an antibody comprises three segments called complementarity-determining regions (“CDR”) or hypervariable regions. Each CDR in a light chain, together with the corresponding CDR in the adjacent heavy chain, form an antigen- binding site of the antibody. Light chains are of two major types, k and l, depending on their constant region. Both k and l light chains may combine with any of the different heavy chain types.
[0006] Amyloid light-chain amyloidosis (AL amyloidosis, AL, or ALA), also called primary amyloidosis, is the most common form of systemic amyloidosis in the United States. The term“amyloidosis” refers to a cluster of diseases which share a common feature, i.e., the extracellular deposition of pathologic insoluble fibrillar proteins in organs and tissues (Rodney, et al. - NEJM, 25:898). Amyloidosis is caused by malfunction of a person’s antibody- producing cells causing production of abnormal protein fibers which aggregate to form insoluble amyloid deposits in organs and tissues. The type of amyloidosis is determined by the nature of the precursor proteins which form the fibril deposit. In primary amyloidosis, the fibrils comprise fragments of immunoglobulin light chains and in secondary amyloidosis, the fibrils comprise amyloid A protein. Modern classification of amyloidosis is based on the nature of the precursor plasma proteins which form the fibril deposit.
[0007] The precursor plasma proteins are diverse and unrelated. Nevertheless, all precursor deposits produce amyloid deposits that share a common typical b-pleated-sheet configuration, which is responsible for the typical staining properties of the fibrillar deposits. The final stage in the development of amyloidosis is the deposit of amyloid fibrils in the organs of the sufferer. Amyloidosis mortality is high, with current five-year survival rates of about 28%.
[0008] To date, the treatment of AL has been directed towards reducing the synthesis of amyloidogenic precursor light chains by attacking the malfunctioning cells through conventional or high dose cytotoxic chemotherapy. Despite the improved prognosis gained by eliminating the offending plasma cell clone, mortality remains high due to multi-organ dysfunction caused by persistent, insoluble amyloid fibril deposits. This treatment suffers from two disadvantages. First, the fibrillar amyloid deposits are often asymptomatic until after significant deposition has taken place. Therefore, treatment is unlikely to be undertaken before significant deposits have already occurred. Second, since this treatment is, at best, effective only to stop the production of precursor abnormal protein but not to remove the existing deposits, prognosis for AL patients remains exceedingly poor due to persistence (or progression) of the pathologic deposits (Solomon, et at. - Int. J. Exp. Clin. Invest. 2:269).
[0009] As a result, therapeutic targeting and clearance of amyloid deposits is an area of intense medical interest. Ancillary to this is intense interest in detecting the presence and location of amyloid deposits and monitoring the clearance of amyloid deposits as an adjunct to the aforementioned targeting and clearance. The compositions and methods disclosed herein fulfill this need for detecting such presence and location.
SUMMARY OF THE INVENTION
[0010] Described herein are compositions and methods for detecting the presence, location, and amount of amyloid deposits, including those resulting from primary (AL) amyloidosis. The disclosed compositions comprise a humanized or chimeric antibody or fragment thereof (“antigen-binding fragment”) that specifically binds to amyloid deposits ( e.g ., amyloid light chain fibrils) and to which is linked a detectable molecule. The disclosed method comprises administering to a subject suspected of having amyloid deposits the above-described composition and detecting the presence, quantity, and/or location of the amyloid deposits by detection of the detectable molecule by diagnostic imaging.
[0011] The present methods also include a method of stratifying patients for therapy with the disclosed humanized or chimeric antibody based on the affinity of the diagnostic composition for the amyloid deposits. The present methods also include determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy based on the affinity of the diagnostic compositions for the amyloid deposits. That is, one may determine the appropriate dosing of a patient based on the detected affinity (uptake) of the labeled antibody or antibody fragment. A patient showing strong affinity for the labeled antibody or antigen-binding antibody fragment may require a smaller amount of therapeutic antibody or antigen-binding antibody fragment than a patient showing a weaker affinity for the labeled antibody or antigen-binding antibody fragment. The present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the affinity of the labeled antibody or antibody fragment for the amyloid deposits, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment based on the strength of the affinity.
[0012] In some embodiments, the disclosed antibody in the diagnostic compositions comprises a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48. In some embodiments, the antibody comprises a constant region derived from a human lgG1. In some embodiments, the antibody binds to amyloid fibrils with a higher affinity than its murine equivalent. In some embodiments, the antibody binds to an epitope expressed by the b-pleated sheet configuration of amyloid fibrils with higher affinity than a mouse antibody comprising a VK region of SEQ ID NO: 36 and a VH region of SEQ ID NO: 35. And in some embodiments, the antibody binds to kappa and lambda amyloid fibrils in vivo.
[0013] In another aspect, the present disclosure provides compositions comprising the disclosed humanized or chimeric antibody linked to a detectable label or molecule and a pharmaceutically acceptable carrier. [0014] A chimeric antibody useful in the subject methods and compositions may be produced by co-transfection in mammalian cells of the vector constructs 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200 or transfection in mammalian cells of the supervector construct pG1 KD200-11-1 F4. In some embodiments, the co-transfection of the vector constructs 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200 or transfection of a supervector construct pG1 KD200- 11-1 F4 takes place in COS cells. The antibody thus produced is designated “chimeric 11-1 F4 antibody”. As disclosed elsewhere herein, fragments of the chimeric 11-1 F4 antibody are also useful in the subject methods and compositions.
[0015] In some embodiments, the amyloid deposits detected result from primary amyloidosis. In some embodiments, the primary amyloidosis comprises involvement of at least one organ or tissue selected from the group consisting of heart, kidneys, liver, lung, gastrointestinal tract, nervous system, muscular skeletal system, soft tissue, and skin and the deposits are detected in one or more of those organs.
[0016] In one aspect, the present disclosure provides methods of detecting or imaging amyloid deposits comprising administering to a patient diagnosed with or suspected of having an amyloid deposition disease a diagnostically effective amount of a humanized or chimeric antibody or an antigen-binding fragment thereof having a detectable label or marker linked thereto and detecting the presence, location, and/or quantity of the detectable label bound to amyloid deposits in the patient’s body by diagnostic imaging. In this aspect, the antibody or antigen-binding fragment may comprise: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 52; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51. In another aspect, the present disclosure provides compositions of the aforementioned antibody or antibody fragment linked to a detectable molecule and a pharmaceutically-acceptable carrier. [0017] In some embodiments of the foregoing aspects, the antibody or antigen-binding fragment thereof may be a humanized antibody, while in some embodiments, the antibody or antigen-binding fragment thereof may be a chimeric antibody.
[0018] In some embodiments of the foregoing aspects, the VK region of the antibody or antigen-binding fragment may comprise SEQ ID NO: 47 and the VH region may comprise SEQ ID NO: 48.
[0019] In some embodiments of the foregoing aspects, the antibody or antigen-binding fragment may comprise a constant region that is derived from a human lgG1. In some embodiments, the antibody may be chimeric 11 -1 F4 antibody.
[0020] In another aspect, the present disclosure provides methods of detection of an amyloid deposition disease in a patient suspected of having such disease by administering a labeled antibody or an antigen-binding fragment thereof and detecting the presence of the label in the patient by diagnostic imaging.
[0021] In some embodiments of this aspect, the humanized or chimeric 11-1 F4 antibody comprises a constant region is derived from a human lgG1.
[0022] In another embodiment, the present disclosure provides an in vivo method of detecting the presence, location, and/or amount of amyloid deposits in a patient suspected of having amyloid deposits which comprises administering to the patient an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51 ; and detecting the presence, location, and/or quantity of amyloid deposits by detection of the detectable label bound to the amyloid deposits by diagnostic imaging.
[0023] In another embodiment, the disclosure provides an in vivo method of detecting the presence, location, and/or quantity of amyloid deposits in a patient suspected of having amyloid deposits which comprises administering to the patient an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48; and detecting the presence, location, and/or quantity of amyloid deposits bound to the amyloid deposits by detection of the detectable label by diagnostic imaging.
[0024] In one aspect of either of the above methods, the disclosure provides the above-described methods wherein the method of detection is positron emission spectroscopy (PET). In the aspects wherein the method of detection is PET, the detectable label may be 124l or 89Zr. In another aspect of the above methods, the antibody is selected from a chimeric or humanized 11 - 1 F4, antigen-binding fragments thereof, and the antibody CAEL-101. In still another aspect of the above methods, the amyloid deposits are in the heart.
[0025] In another embodiment, the present disclosure provides a composition for detecting the presence of amyloid deposits in a subject which comprises an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51.
[0026] In another embodiment, the present disclosure provides a composition for detecting the presence of amyloid deposits in a subject which comprises an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising: a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
[0027] In one aspect of the above compositions, the detectable molecule is selected from 124l and 89Zr. In another aspect of the above compositions, the antibody is selected from chimeric or humanized 1 1 -1 F4 or antigen-binding fragments thereof and the antibody CAEL-101 . In yet another aspect of the above compositions, the amyloid deposits are in the heart.
[0028] In another embodiment, the present disclosure provides a method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprising the steps of: a) administering to said patient a composition described in the previous paragraphs and conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, b) treating the patient with a therapy intended to remove amyloid deposits, c) administering to said patient a composition described in the previous paragraphs, d) conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, and e) comparing the detected amount of detectable molecule in step d to the detected amount of detectable molecule in step a.
[0029] In the above method, the patient may be treated in step b) with an antibody selected from a humanized or chimeric 11 -1 F4 antibody or antigen-binding fragment thereof and the antibody CAEL-101 . In the above method, the composition may be any of the compositions described above. In another aspect, the method of detection may be PET. In another aspect, if the detection method is PET, the detectable label is selected from 124l and 89Zr. In another aspect of the above methods, the antibody is selected from humanized or chimeric 1 1 -1 F4 or antigen-binding fragments thereof and CAEL-101 . In another aspect of the above methods, the amyloid deposits are in the heart. [0030] In another embodiment, the present disclosure provides a method of determining the effectiveness of treatment to remove amyloid deposits in a patient comprising a) treating the patient with a therapeutically- effective dose of the humanized or chimeric 11-F4 antibody or antibody fragment thereof, b) administering to the patient a diagnostically-effective amount of one of the above-described compositions, and c) measuring by diagnostic imaging the amount of detectable molecule from the diagnostic composition in the lymph nodes of the patient, wherein the higher the amount of detectable molecule detected in the lymph nodes, the more effective the treatment. In one aspect of this embodiment, the amyloid deposits are in the heart.
[0031] In some embodiments of this disclosure, the chimeric 11-1 F4 antibody is CAEL-101 , the antibody produced by the CHO cells deposited with the ATCC as ACC No. PTA-125146.
[0032] In some embodiments, the primary amyloidosis consists of lambda light chain fibril aggregate deposits, while in some embodiments, the primary amyloidosis consists of kappa light chain fibril aggregate deposits, and in still other embodiments, the primary amyloidosis consists of kappa and lambda light chain fibril aggregate deposits.
[0033] The foregoing general description and following detailed description are exemplary and explanatory and are intended to provide further explanation of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0034] Figure 1 outlines the strategy used to clone the murine VH and VK genes from a hybridoma cell line.
[0035] Figure 2 is a listing of DNA and amino acid sequences of the murine 11-1 F4 antibody VH region gene, SEQ ID NO: 39 and NO: 35, respectively. [0036] Figure 3 is a listing of DNA and amino acid sequences of the murine 11-1 F4 antibody VK region gene, SEQ ID NO: 40 and NO: 36, respectively.
[0037] Figure 4 is a map of the immunoglobulin kappa light chain expression vector pKN100. It consists of a pSV2 vector fragment, which has the SV40 early and crippled SV40 late promoter, the SV40 origin and the Co1 E1 origin. It also has the ampicillin resistance and neo genes. The crippled SV40 late promoter drives the neo genes. It also has the HCMVi promoter, a multiple cloning site (containing the BamH\ and Hind III restriction sites) for the insertion of an immunoglobulin variable region gene, and cDNA for the human kappa constant region gene terminated by a spaC2 termination signal sequence ("Arnie"), which is in the same orientation as the kappa light chain expression cassette.
[0038] Figure 5 is a map of the immunoglobulin gamma 1 heavy chain expression vector pG1 D200. It consists of a pSV2dhfr vector fragment, which has the SV40 early and crippled SV40 late promoter, the SV40 origin, and the Co1 E1 origin. It also has the ampicillin resistance and dhfr genes. The crippled SV40 late promoter drives the dhfr gene. Consequently, expression is poor, allowing for the selection of multigene/high expression level clones using comparatively low levels of methotrexate. It also has the HCMVi promoter fragment, a multiple cloning site, cDNA for a human gamma 1 constant region gene (intron minus) which is followed by a spaC2 termination signal sequence ("Amie").
[0039] Figure 6 is a listing of the DNA and amino acid sequences of the modified murine 11-1 F4 antibody VK region gene (SEQ ID NO: 42 and NO: 47, respectively) and the sequences of the oligonucleotide primers used to modify the VK gene (SEQ ID NO: 41 and NO: 43, respectively).
[0040] Figure 7 is a listing of the DNA and amino acid sequences of the modified murine 11-1 F4 antibody VH region gene (SEQ ID NO: 45 and NO: 48, respectively) and the sequences of the oligonucleotide primers used to modify the VH gene (SEQ ID NO: 44 and NO: 46, respectively).
[0041] Figure 8 is a graphical representation of the result of the amyloid fibril binding ELISA assay. The cos cell supernatants containing chimeric 11 - 1 F4 antibody were tested separately on the same ELISA plate along with purified murine 11-1 F4 antibody. The absorbance was read at OD405. New sv = pG1 KD200-11-1 F4. New co-transfection = 11-1 F4VHpG1 D200 plus 11- 1 F4VK.pKN100.
[0042] Figure 9 is a PET image of a mouse implanted with human cardiac derived amyloidoma which was injected with [124I]CAEL-101 and imaged at day 4 after injection.
DETAILED DESCRIPTION OF THE INVENTION
[0043] In accordance with the present disclosure, compositions comprising humanized antibodies, chimeric antibodies ( e.g ., mouse-human antibodies) or antigen-binding fragments thereof to which in each case is linked a detectable molecule are provided that are useful for detecting the presence, location, and amount of amyloid deposits in vivo. These compositions allow detection, localization, and/or quantification of amyloid deposits while producing little or no human anti-mouse antibody (HAMA) reaction. The disclosure provides compositions comprising at least one of said antibodies or antibody fragments linked to a detectable marker and a pharmaceutically acceptable carrier and methods of detecting the location and amount of amyloid deposits by administering to a patent an amount of said marker-linked antibody or antibody fragment effective to allow detection of amyloid deposits if such deposits are present and detecting the presence, amount, and/or location of the detectable marker bound to the amyloid deposits by diagnostic imaging.
[0044] Further in accordance with the present disclosure, methods are provided for monitoring or determining the effect of treating amyloid deposition diseases. The method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprises the steps of:
a. administering to said patient a diagnostically effective amount of a humanized or chimeric 11 -1 F4 antibody or an antigen-binding fragment thereof having a detectable molecule linked thereto and detecting the amount of detectable molecule bound to the amyloid deposits,
b. treating the patient with therapy intended to remove amyloid deposits, such as a humanized or chimeric 11-1 F4 antibody or antigen-binding fragment thereof,
c. administering to said patient a diagnostically effective amount of a humanized or chimeric 11 -1 F4 antibody or an antigen-binding fragment thereof having a detectable molecule linked thereto and detecting the amount of detectable molecule bound to the amyloid deposits, and
d. comparing the detected amount of detectable molecule in step c to the detected amount of detectable molecule in step a.
[0045] In this embodiment, the present disclosure is directed towards measuring the extent of change in the amount of amyloid deposits of a patient (if any) by administering to said patient the marker-linked antibody or antibody fragment as described herein both prior to and after a treatment intended to remove amyloid deposits and measuring the difference in the size and extent of the detected amyloid deposits after the treatment compared to the size and extent measured prior to the treatment.
[0046] Further provided is an in vivo method of detecting amyloid deposits in a subject, the method comprising: a) administering to the subject a composition comprising one or more than one antibody or antigen-binding fragment thereof as described herein linked to a detectable molecule; and b) detecting by diagnostic imaging the detectable agent linked to the antibody or antigen-binding fragment thereof bound to the amyloid deposits.
[0047] In the method described above, the step of detecting (step b) may be performed using PET (positron emission tomography), SPECT (single- photon emission computed tomography), MRI, fluorescence imaging, or any other suitable diagnostic imaging method. PET is the preferred imaging method. The use of PET for imaging tumors or other deposits using a detectable molecule is well known. This preparation of diagnostic imaging agents and their use in PET are described, for example, in Bailly, et al. - Int. J. Mol. Sci. 2017, 18, 57 ; Boerman, et al. -J Nucl Med, 2011; 52:1171-1172] and Mayer, et al. - J Nucl Med 2017; 58:538-546, all of which are incorporated herein by reference in their entirety. Although these references are directed mainly at cancerous tumor detection, the materials and methods disclosed therein are equally useful for detecting amyloid deposits.
[0048] The in vivo imaging step in the methods described above may be whole body imaging for diagnostic purposes or local imaging at specific sites, such as but not limited to heart, kidney, liver, spleen, nervous system, or digestive system in a quantitative manner to assess the progression of disease or patient response to a treatment regimen. The detection step in the methods as described above may be any diagnostic imaging technology including, but not limited to: positron emission tomography (PET), wherein the detectable agent is an isotope such as 11C, 13N, 150, 18F, 64Cu, 62Cu, 124l, 76Br, 82Rb, 89Zr, or 68Ga; single photon emission computed tomography (SPECT), wherein the detectable agent is a radiotracer such as 99mTc, 1111n, 123l, 201TI, or 133Xe, depending on the specific application; magnetic resonance imaging (MRI), wherein the detectable agent may be, for example and not limited to, gadolinium, iron oxide nanoparticles and carbon-coated iron-cobalt nanoparticles. In illustrative examples herein, antibodies are labeled with 124l or 89Zr. [0049] The detectable molecule (sometimes described herein as a “label”, “marker”, or “agent”) as described herein may be linked, also sometimes referred to herein as "conjugated", to the antibody or fragment thereof by any suitable method known in the art. For example, and without wishing to be limiting, the detectable agent may be linked to the antibody or antibody fragment by a covalent bond or ionic interaction. The linkage may be achieved through a chemical cross-linking reaction, or through fusion using recombinant DNA methodology combined with any peptide expression system, such as bacteria, yeast or mammalian cell-based systems. Methods for linking an antibody or fragment thereof to a detectable agent are well-known to a person of skill in the art, as shown in the references cited above.
[0050] Further provided herein is a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy based on the affinity of the diagnostic compositions for the amyloid deposits as determined by the uptake of the diagnostic composition by the amyloid deposits of the patient. That is, one may determine the dosing of a patient based on the detected uptake of the labeled antibody or antibody fragment. A patient showing higher uptake of the labeled antibody or antibody fragment may require a smaller amount of therapeutic antibody or antibody fragment than a patient showing a lower uptake of the labeled antibody or antibody fragment. The present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the uptake of the labeled antibody or antibody fragment by the amyloid deposits, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment based on the detected amount of uptake.
[0051] Further provided herein is a method of determining the effectiveness of a treatment with the chimeric or humanized antibody or antibody fragment disclosed herein to remove amyloid deposits. The method comprises: a) treating the patient with a therapeutically-effective dose of a therapy intended to remove amyloid deposits, such as the chimeric or humanized antibody or antibody fragment disclosed herein, b) administering to the patient a diagnostically-effective amount of the diagnostic composition disclosed herein, and c) measuring the amount of detectable molecule from the diagnostic composition in the lymph nodes of the patient, wherein the higher the amount of detectable molecule detected in the lymph nodes, the more effective the treatment.
Definitions
[0052] It is to be understood that methods are not limited to the particular embodiments described, and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. The scope of the present technology will be limited only by the appended claims.
[0053] As used herein, certain terms may have the following defined meanings. As used in the specification and claims, the singular form“a,”“an” and “the” include singular and plural references unless the context clearly dictates otherwise. For example, the term“a cell” includes a single cell as well as a plurality of cells, including mixtures thereof.
[0054] As used herein, the term“comprising” is intended to mean that the compositions and methods include the recited elements, but not excluding others. “Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the composition or method. “Consisting of shall mean excluding more than trace elements of other ingredients for claimed compositions and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. Accordingly, it is intended that the methods and compositions can include additional steps and components (comprising) or alternatively including steps and compositions of no significance (consisting essentially of) or alternatively, intending only the stated method steps or compositions (consisting of). [0055] As used herein,“about” means plus or minus 10%.
[0056] As used herein, “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
[0057] As used herein, the terms“individual”,“patient”, or“subject” can be an individual organism, a vertebrate, a mammal ( e.g ., a bovine, a canine, a feline, or an equine), or a human. In a preferred embodiment, the individual, patient, or subject is a human.
[0058] As used herein, the term an“isolated antibody” is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities {e.g., an isolated antibody that specifically binds to an amyloid fibril is substantially free of antibodies that do not bind to amyloid fibrils). An isolated antibody that specifically binds to an epitope of an amyloid light chain fibril {e.g., a kappa and/or lambda fibril) may, however, have cross- reactivity to other proteins, such as amyloid A fibrils. However, the antibody preferably always binds to human amyloid light chain fibrils. In addition, an isolated antibody is typically substantially free of other cellular material and/or chemicals.
[0059] As used herein, the phrase “diagnostically effective amount” means the amount of detectable marker-linked antibody or antibody fragment which, when administered to a patient or subject, permits in vivo detection, localization, and/or quantification by diagnostic imaging of amyloid deposits in the patient or subject if any such deposits are present. It is emphasized that a diagnostically effective amount will not always be effective to detect amyloid deposits, even though such amount is deemed to be a diagnostically effective amount by those of skill in the art. The diagnostically effective amount may vary based on the route of administration and dosage form, the age and weight of the subject, and/or the subject’s condition, including the type and stage of the amyloidosis at the time that diagnosis commences, among other factors.
[0060] As used herein, the term “humanized antibody” refers to an antibody that comprises the CDRs of antibodies derived from mammals other than human, and the framework region (FR) and the constant region of a human antibody. A humanized antibody is useful as a component in a diagnostic composition according to the present disclosure since antigenicity of the humanized antibody in human body is lowered.
[0061] As used herein, the term“antibody fragment” refers to a part of an antibody that comprises a CDR of the antibody but has been engineered to delete some of the structure of the intact antibody. Engineering antibody fragments is well known in the art, as shown (for example) in Holliger, et al. - Nature Biotechnology: 23 (9), 1126-1135 (2005), incorporated herein by reference in its entirety. The term“antigen-binding fragment” as used herein means an antibody fragment that binds to amyloid deposits.
[0062] As used herein, the term “pharmaceutically-acceptable carrier” means a material for admixture with a pharmaceutical or diagnostic compound (e.g., a chimeric antibody linked to a detectable molecule) for administration to a patient as described, for example, in“Ansel’s Pharmaceutical Dosage Forms and Delivery Systems”, Tenth Edition (2014).
[0063] As used herein, the synonymous terms “detectable molecule”,
“detectable marker”, “detectable agent”, and “detectable label” refer to substances that may be detected in vivo by diagnostic imaging techniques, such as (but not limited to) PET, SPECT, and MRI.
[0064] As used herein, the terms“diagnostic imaging” or“imaging” mean methods of in vivo imaging of a detectable molecule in a patient or subject to determine the presence, location and/or amount of a tissue or substance (such as an amyloid deposit) in the patient or subject to which the detectable molecule has been localized, such as by being linked to an antibody or antibody fragment that binds to the tissue or substance to be detected. Anti-AL Antibodies
Murine anti-fibril antibodies
[0065] Recent animal studies have shown that the administration of the murine 11-1 F4 antibody and other murine anti-human light chain specific antibodies directed against an epitope common to the b-pleated-sheet structure present on AL fibrils results in complete degradation of the human ALK and ALA amyloid deposits. Some of these murine antibodies are described in United States patent 8,105,594 (“the ‘594 patent”), which is incorporated herein by reference in its entirety.
[0066] Murine antibodies are generally unsuitable for administration to other animal species (such humans) because the receiving species will recognize the murine antibody as antigenic and will produce antibodies against it. The antigenicity of an antibody from one species when injected into another species is normally caused by a portion of a constant domain. Such an antigenic response will impede or prevent the desired therapeutic or diagnostic effect of the murine antibody. In humans, this antigenic response is called human anti-mouse antibody (HAMA). The antibodies described in the‘594 patent have the potential to be highly immunogenic in humans via the human anti-mouse antibody (HAMA) response. Since the HAMA response usually results in the rapid clearance of a mouse antibody from the human recipient, HAMA would severely limit any potential human therapeutic or diagnostic benefit a murine antibody could have. Therefore, these murine antibodies are unsuitable for administration to a patient to detect deposits of amyloid fibrils in a patient. Thus, the present disclosure provides compositions and methods for detecting and/or monitoring amyloid deposition diseases that is less likely to produce an immunogenic HAMA response in a patient following administration.
Humanized and chimeric anti-fibril antibodies
[0067] The present disclosure provides compositions comprising humanized and chimeric antibodies or antigen-binding fragments thereof having linked thereto a detectable molecule. The disclosed compositions are useful for imaging the location and amount of amyloid deposits in a subject. Typically, an antibody consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two copies of a light (L) chain polypeptide. Typically, each heavy chain contains one N-terminal variable (VH) region and three C-terminal constant (CH1 , CH2 and CH3) regions, and each light chain contains one N-terminal variable (VL or VK) region and one C-terminal constant (CL) region. Each variable domain of the light and heavy chain in an antibody also comprises three segments called complementarity-determining regions (“CDR”) or hypervariable regions. Each CDR in a light chain, together with the corresponding CDR in the adjacent heavy chain, form an antigen-binding site of the antibody. The variable regions of each pair of light and heavy chains form the antigen binding site of an antibody, whereas the constant region provides structural support and modulates the immune response initiated by the antigen binding.
[0068] Chimeric antibodies incorporate the variable region of a non-human antibody into the constant region of a human antibody. A chimeric 11-1 F4 antibody, for instance, may be created by expressing the murine variable region with the Fc region of a human antibody, such as a human lgG1.
[0069] Humanized forms of non-human ( e.g ., murine) antibodies can be obtained, which contain minimal sequences derived from non-human immunoglobulin. In general, a humanized antibody may comprise one or two or more variable domains in which variable regions are derived from non-human immunoglobulin and framework regions (FR) correspond to a human immunoglobulin sequence. Thus, in some embodiments, a humanized anti-AL antibody comprises a human antibody framework region. Such antibodies can be prepared by known techniques.
[0070] The murine 11-1 F4 monoclonal antibody is an anti-AL antibody produced by the SP2/0 hybridoma cell deposited by Alan Solomon, MD (University of Tennessee Medical Center at Knoxville, TN). The hybridoma cell line is available from the American Type Culture Collection (ATCC access PTA- 105). The VK region (SEQ ID NO: 36) and the VH region (SEQ ID NO: 35) of the 11-1 F4 antibody are shown in Table 1 below. The CDR sequences for the heavy and light chains and provided in Table 2.
Table 1 -11-1 F4 monoclonal antibody variable sequences
Figure imgf000021_0002
_
Table 2 -11-1 F4 monoclonal antibody CDR sequences
Figure imgf000021_0001
Figure imgf000022_0001
[0071] One can clone the genes for the Vhi and VK regions shown above to produce a chimeric 11-1 F4 antibody using known human antibody sequences. The chimeric 11-1 F4 antibody binds to an epitope expressed by the b-pleated sheet configuration of amyloids, just as its murine counterpart does, but surprisingly, as shown in Example 6 below, the chimeric antibody binds to AL amyloid fibrils with higher affinity than the 11-1 F4 mouse antibody from which it was derived.
[0072] One can also clone the genes for the CDR regions to produce a humanized form of the antibody using known human antibody sequences. Like the chimeric form of the 11-1 F4 antibody, the humanized form may also have a binding affinity for amyloid fibrils that is higher than that of the murine counterpart.
[0073] Those of skill in the art will understand that the disclosed humanized and chimeric antibodies may utilize all different types of human constant regions and/or framework regions. For example, the disclosed humanized and chimeric antibodies may comprise the constant regions and/or framework regions of a human IgG (including lgG1 , lgG2, lgG3, and lgG4), IgA, IgE, IgH, or IgM. In preferred embodiments, the disclosed humanized or chimeric 11-1 F4 antibody comprises a human lgG1 constant region.
[0074] In some embodiments, the disclosed antibodies may comprise one or more substitutions, insertions, or deletions, so long as the antibody maintains the ability to bind to amyloid fibrils ( e.g ., kappa and/or lambda light chain fibrils). For example, in some embodiments, a chimeric 11 -1 F4 antibody of the present disclose may comprise heavy and light chains with about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91 %, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% identity compared to the corresponding heavy and light chain sequences disclosed herein, so long as the antibody maintains the ability to bind to amyloid fibrils. In some embodiments, a humanized 11 -1 F4 antibody of the present disclose may comprise CDRs that have about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91 %, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% identity compared to the corresponding CDR sequences disclosed herein, so long as the antibody maintains the ability to bind to amyloid fibrils.
Abbreviations
[0075] Dulbecco's Modified Eagles Medium (DMEM), Fetal Bovine Serum (FBS), ribonucleic acid (RNA); messenger RNA (mRNA); deoxyribonucleic acid (DNA); copy DNA (cDNA); polymerase chain reaction (PCR); minute (min); second (sec); Tris-borate buffer (TBE).
[0076] Amino acids are represented by the IUPAC abbreviations, as follows: Alanine (Ala), Arginine (Arg), Asparagine (Asn), Aspartic acid (Asp), Cysteine (Cys), Glutamine (Gin), Glutamic acid (Glu), Glycine (Gly), Histidine (His), Isoleucine (lie), Leucine (Leu), Lysine (Lys), Methionine (Met), Phenylalanine (Phe), Proline (Pro), Serine (Ser), Threonine (Thr), Tryptophan (Trp), Tyrosine (Tyr), Valine (Val). Similarly for nucleotides: Adenine (A), Cytosine (C), Guanine (G), Thymine (T), Uracil (U), Adenine or Guanine (R), Cytosine or Thymine (Y), Guanine or Cytosine (S), Adenine or Thymine (W), Guanine or Thymine (K), Adenine or Cytosine (M), Cytosine or Guanine or Thymine (B), Adenine or Guanine or Thymine (D), Adenine or Cytosine or Thymine (H), Adenine or Cytosine or Guanine (V), and any base (N).
Flumanized or Chimeric Antibodies
[0077] To produce the chimeric antibodies of the invention, the murine 11-1 F4 monoclonal antibody heavy and kappa light chain variable region genes described in United States patent 8,105,594 were PCR modified to facilitate the expression of the chimeric 11-1 F4 antibody in mammalian cells.
A detailed sequence analysis of the modified variable region genes was performed. The modified variable region genes were cloned into the appropriate mammalian expression vectors, creating the constructs 11 - 1 F4VHpG1 D200 and 11 -1 F4VK.pKN100. A single supervector construct, pG1 KD200-11-1 F4, was made from the 11-1 F4VHpG1 D200 and 11 - IF4VK.pKN100 constructs by EcoRI restriction enzyme digest and ligation. Finally, the chimeric 11 -1 F4 antibody was transiently expressed in COS cells by both cotransfection and single supervector transfection. While COS cells were chosen for the co-transfection or transfection as a matter of convenience, those of skill in the art would recognize that other mammalian cell lines could be used. The characterization of the binding capacity of the chimeric 11-1 F4 antibody for amyloid fibrils was determined by direct binding ELISA. Unexpectedly and beneficially, the chimeric 11-1 F4 antibody bound to amyloid fibrils with higher affinity than the murine 11 -1 F4 antibody.
[0078] Typically, an antibody consists of four polypeptides: two identical copies of a heavy (FI) chain polypeptide and two copies of a light (L) chain polypeptide. Typically, each heavy chain contains one N-terminal variable (VH) region and three C-terminal constant (CH1 , CH2 and CH3) regions, and each light chain contains one N-terminal variable (VL or VK) region and one C- terminal constant (CL) region. The variable regions of each pair of light and heavy chains form the antigen binding site of an antibody.
[0079] An antibody useful in the compositions and methods of the invention may be a chimeric mouse-human monoclonal antibody comprising the VK region of SEQ ID NO: 47 and the VH region of SEQ ID NO: 48 or a humanized monoclonal antibody comprising CDR sequences of SEQ ID NOs: 49-54. These antibodies bind to an epitope expressed by the b-pleated sheet configuration of amyloid fibrils. Moreover, surprisingly the antibodies bind to this epitope with higher affinity than the 11-1 F4 mouse antibody from which they were derived, which comprises the VK region of SEQ ID NO: 36 and the VH region of SEQ ID NO: 35. The invention includes methods of detecting amyloid deposits in a human patient which comprises administering to the patient a diagnostically effective dose of one of the above antibodies linked to a detectable molecule in a pharmaceutically-acceptable carrier. The antibody composition may be administered by any conventional route of administration, but parenteral administration (such as intravenous) is preferred. Pharmaceutically-acceptable carriers are well-known in the art and a suitable one can be selected by one of skill in the medical field.
[0080] Chimeric antibodies and antigen binding fragments thereof useful for the compositions and methods described and claimed herein (and a method of making the chimeric antibodies) are disclosed in co-owned Patent Cooperation Treaty application PCT/US18/399805 (docket 8441-0004WO with priority to United States patent application 62/526,835, filed June 29, 2017), filed June 28, 2018, and co-owned Patent Cooperation Treaty Application PCT/US2018/043374 (docket 8441-0009WO) filed July 24, 2018, both of which are incorporated herein in its entirety. Materials useful to make the subject antibody include vector constructs selected from the group consisting of 11-1 F4VK.pKN100 and 11-F4VH.pG1 D200, shown in Figures 5 and 6, respectively, and the superconstruct pG.1 KD20011-1 F4 made from the two above vector constructs. Other useful materials include the modified murine 11-1 F4 antibody VK region gene (SEQ ID NO: 42) and the modified 11- 1 F4 antibody VH region gene (SEQ ID NO: 45), as well as the respective primers SEQ ID NO: 41 , 43, 44, and 46. The subject antibody may be made by co-transfection of the vector constructs 11 -1 F4VK.pKN100 and 11 - F4VFI.pG1 D200 or the superconstruct pG.1 KD20011 -1 F4 in a suitable mammalian host cell, such as COS (Chinese hamster ovary) cells.
[0081] Methods of making, testing, and using the humanized or chimeric 11-1 F4 antibody are discussed in further detail in the Examples section below.
Diagnostic Formulations
[0082] Diagnostic compositions suitable for use in the methods described herein can include the disclosed humanized or chimeric 11-1 F4 antibodies, humanized antibodies, or antigen-binding antibody fragments linked in each case to a detectable marker, and a pharmaceutically acceptable carrier or diluent. [0083] The composition may be formulated for intravenous, subcutaneous, intraperitoneal, or intramuscular administration, although intravenous administration is preferred.
[0084] Pharmacologically acceptable carriers for various dosage forms are known in the art. For example, solvents, solubilizing agents, suspending agents, isotonicity agents, buffers, and soothing agents for liquid preparations are known. In some embodiments, the pharmaceutical compositions include one or more additional components, such as one or more preservatives, antioxidants, stabilizing agents and the like.
[0085] Sterile injectable solutions can be prepared by incorporating the marker-linked antibody or antibody fragment in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the diagnostic composition into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
Methods of Diagnosis and Monitoring
[0086] In general, amyloidosis is caused by the buildup of an abnormal protein called amyloid. Amyloid is produced in the bone marrow and can be deposited in any tissue or organ. The specific cause of the condition depends on the type of amyloidosis.
[0087] There are several types of amyloidosis or amyloid diseases, including AL amyloidosis, AA amyloidosis, and hereditary amyloidosis.
[0088] AL amyloidosis (immunoglobulin light chain amyloidosis) is the most common type and can affect the heart, kidneys, skin, nerves and liver. Previously known as primary amyloidosis, AL amyloidosis occurs when the bone marrow produces abnormal antibodies that cannot be broken down. The antibodies are deposited in various tissues as amyloid plaques (amyloid deposits), which interfere with normal function of the tissue or organ. [0089] AA amyloidosis generally affects the kidneys but occasionally also affects the digestive tract, liver or heart. It was previously known as secondary amyloidosis. It often occurs along with chronic infectious or inflammatory diseases, such as rheumatoid arthritis or inflammatory bowel disease.
[0090] Hereditary amyloidosis (familial amyloidosis) is an inherited disorder that usually often affects the liver, nerves, heart, and/or kidneys. Many different types of gene abnormalities present at birth are associated with an increased risk of amyloid disease or hereditary amyloidosis. The type and location of an amyloid gene abnormality can affect the risk of certain complications, the age at which symptoms first appear, and the way the disease progresses over time.
[0091] When an amyloid disease affects the heart, it can cause numerous types of complications. Amyloid deposits or plaques reduce the heart’s ability to fill with blood between heartbeats. Less blood is pumped with each beat, and this may lead to shortness of breath. Amyloid deposits or plaques in or around the heart may also cause irregular heartbeats and congestive heart failure, among other organ dysfunctions
[0092] When an amyloid disease affects the kidneys, it will often harm the kidneys’ filtration ability, allowing protein to leak from the blood into the urine (i.e., proteinuria). Moreover, the kidneys’ ability to remove waste products from your body is lowered, which may eventually lead to kidney failure.
[0093] Provided herein are methods of detecting, locating, and/or quantifying amyloid deposits in a patient suffering or suspected to be suffering from an amyloid deposition disease, such as primary (AL) amyloidosis, by administering to the patient ( e.g ., a human patient) a humanized or chimeric 11-1 F4 antibody or an antigen-binding fragment thereof linked to a detectable marker together with a pharmaceutically acceptable carrier, in an amount effective to detect, locate, and/or quantify amyloid deposits if such are present, followed by detecting the detectable molecule bound to the amyloid deposits in the patient by diagnostic imaging to determine the extent, location, and/or amount of the amyloid deposits if such are present.
[0094] Also provided herein is a method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprising the steps of:
a. administering to said patient a diagnostically effective amount of a humanized or chimeric 11-1 F4 antibody or an antigen- binding fragment thereof having a detectable molecule linked thereto and conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits,
b. treating the patient with a therapy intended to remove amyloid deposits, such as a humanized or chimeric 11 -1 F4 antibody or antigen-binding fragment thereof,
c. administering to said patient a diagnostically effective amount of a humanized or chimeric 11-1 F4 antibody or an antigen-binding fragment thereof having a detectable molecule linked thereto and conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, and
d. comparing the detected amount of detectable molecule in step c to the detected amount of detectable molecule in step a.
[0095] The above monitoring method can be conducted starting before any treatment has taken place or can be used in the midst of a treatment regimen. That is, step a) above can be performed prior to any treatment of the patient to determine a baseline amount of amyloid deposit or step a) above can be conducted after one or more courses of treatment to determine the effect of the next succeeding course of treatment. [0096] In some embodiments, the amyloid deposition disease ( e.g ., primary amyloidosis) comprises involvement of at least one organ or tissue selected from the group consisting of heart, kidneys, liver, lung, gastrointestinal tract, nervous system, muscular skeletal system, soft tissue, and skin.
[0097] In some embodiments, the disclosed methods comprise monitoring the treatment of a patient suffering from relapse or refractory ALA. In some embodiments, the patient may have kappa ALA. In some embodiments, the patient may have lambda ALA.
[0098] Exemplary diagnostic amounts can vary according to the size and health of the individual being treated, as well as the condition being diagnosed. In some embodiments, the diagnostically effective amount of a radiolabeled humanized or chimeric 11 -1 F4 antibody for PET is about 1-10 mCi. However, in some situations and for other diagnostic imaging modalities the dose may be higher or lower. One of skill in the diagnostic imaging art would know how to select an appropriate amount of labeled antibody or antibody fragment, as described (for example) in the references listed above.
[0099] The following examples are given to illustrate the present invention. It should be understood, however, that the invention is not to be limited to the specific conditions or details described in these examples. All printed publications referenced herein are specifically incorporated by reference.
EXAMPLE 1
PCR Cloning and DNA Sequencing of the mouse 11 -1 F4 Antibody
[00100] The murine 11-1 F4 monoclonal antibody heavy and light chain variable region genes were PCR cloned and a detailed sequence analysis of all variable region genes isolated (both pseudo and functional) was performed. Detailed DNA and amino acid sequences of the murine 11-1 F4 antibody heavy and light chain variable region genes were obtained. Materials
[00101] Media components and all other tissue culture materials were obtained from Life Technologies (UK). The RNA solution kit was obtained from Stratagene (USA), while the first strand cDNA synthesis kit was purchased from Pharmacia (UK). All the constituents and equipment for the RCR-reaction, including AmpliTaq® DNA polymerase, were purchased from Perkin Elmer (USA). The TOPO TA Cloning ® kit was obtained from Invitrogen (USA). Agarose (UltraPure™) was obtained from Life Technologies (UK). The ABI PRISM® Big Dye™ terminator cycle sequencing ready reaction kit pre-mixed cycle sequencing kit and the ABI PRISM® 310 sequencing machine were both purchased from PE Applied Biosystems (USA). All other molecular biological products were obtained from New England Biolabs (USA) and Promega (USA).
Methods
[00102] The strategy used to PCR clone the murine VH and VK genes from the hybridoma cell lines producing the murine monoclonal antibody 11 - 1 F4 is outlined in Figure 1.
[00103] Two clones (B2C4 and B2D6) of the SP2/0 hybridoma cell line producing the ct-human light chain monoclonal antibody 11 -1 F4, were kindly provided by Alan Solomon, MD (University of Tennessee Medical Center at Knoxville, TN). The hybridoma cell line is available from the American Type Culture Collection (ATCC access PTA-105). The cell lines were cultured using DMEM media supplemented with 20% (v/v) FBS, penicillin/streptomycin and L- Glutamine. Cells were cultured until a total viable cell count of 108 cells was reached.
[00104] The cells were harvested separately from each clone as follows. The mouse hybridoma cell line was grown in suspension in an appropriate culture medium and in sufficient quantities to provide a total viable cell count of about 108 cells. The culture supernatant was harvested and the hybridoma cells pelleted in a bench top centrifuge (250 g, 5 min). The cells were gently re-suspended in 20 ml PBS and a 100 pi aliquot was taken for a viable cell count. The cells in the aliquot were pelleted once more and 200 mI of PBS and 200 mI of trypan blue were added to the 100 mI of cells and mixed gently. Ten mI of this mixture was pipetted into a disposable cell-counting slide and the number of white cells in 9 small squares was counted under a microscope. Blue cells (i.e., dead cells) were not counted. The count process was repeated, the results averaged, and the average results multiplied by 9 x 105 to obtain a viable cell count for the cells in 20ml PBS. Once sufficient cells had been harvested, they were re-suspended in I0 ml of Solution D for RNA isolation (see below, Stratagene RNA Isolation Kit).
[00105] Total RNA was then isolated separately from the cells of each clone using a Stratagene RNA isolation kit, according to the manufacturer’s instructions. One ml of 2 M sodium acetate (pH 4.0) was added to the sample and the contents of the tube were thoroughly mixed by repeatedly inverting the tube. To the tube was added 10.0 ml of phenol (pH 5.3-5.7) and the contents again mixed thoroughly by inversion. To the mixture was added 2.0 ml of chloroform-isoamyl alcohol mixture, the tube was capped and vigorously shaken for 10 seconds, and the tube was incubated in ice for 15 minutes. The sample was transferred to a 50-ml thick-walled, round-bottom centrifuge tube that had been pre-chilled on ice and the tube was spun in a centrifuge at 10,000 x g for 20 minutes at 4°C. Two phases were visible in the tube after centrifugation. The upper, aqueous phase contained the RNA, while the lower phenol phase and interphase contained DNA and proteins. The RNA- containing upper, aqueous phase was transferred to a fresh centrifuge tube and the lower phenol phase was discarded. An equal volume of isopropanol was added to the aqueous phase and the contents mixed by inversion, following which the tube was incubated for 1 hour at -20°C to precipitate the RNA. The tube was spun in a centrifuge at 10,000 x g for 20 minutes at 4°C. After centrifugation, the pellet at the bottom of the tube, which contains the RNA, was removed and the supernatant discarded. The pellet was dissolved in 3.0 ml of solution D, 3.0 ml of isopropanol was added to the tube and the contents mixed well. After incubating the tube for 1 hour at -20°C, it was again spun in a centrifuge at 10,000 x g for 10 minutes at 4°C and the supernatant removed from the tube and discarded. (Note: Up to this point the RNA had been protected from ribonucleases by the presence of guanidine isothiocyanate but was now no longer protected.) The pellet was washed with 75% (v/v) ethanol (DEPC-treated water (25%)) and the pellet was dried under vacuum for 2-3 minutes. The RNA pellet is re-suspended in 0.5-2 ml of DEPC-treated water.
[00106] Following the manufacturer’s instructions, an Amersham Pharmacia Biotech first strand cDNA synthesis kit was employed to produce a single- stranded DNA copy of the 11-1 F4 hybridoma mRNA using the Not I- d(T)18 primer supplied with the kit. One reaction was performed for each of the two RNA samples isolated, as follows. The components used were: Bulk first strand cDNA reaction mix, Cloned FPLCpure™ Murine Reverse Transcriptase, RNAguard™, BSA, dATP, dCTP, dGTP, and dTTP, 200 mM DIT aqueous solution, Not l-d(T)18 primer: 5' d[AACTGGAAGAATTCGCGGCCGCAGGAAi8]-3', and DEPC treated water.
[00107] Approximately 5 pg of total RNA in 20 pi DEPC water was heated to 65 'C for I0 min and then chilled on ice. The bulk first strand cDNA reaction mix was pipetted gently to obtain a uniform suspension and the reaction set up in a 0.5 ml microcentrifuge tube as below. 20 mI denatured RNA solution, 11 mI Bulk first strand cDNA reaction mix, 1 mI Not l-d(T)18 primer, and 1 ul DTT solution for 33 mI total volume. The reactants were mixed gently by pipetting and incubated 37°C for 1 hour.
[00108] The murine heavy and kappa light chain variable region genes (VH genes and VK genes, respectively) were then PCR amplified from the ssDNA template using the method described by Jones and Bendig (Bio/Technology 9:88).
[00109] Separate PCR reactions were prepared for each of the degenerate leader sequence specific primers (MHVI - MHV12 for VH and MKVI - MKV11 for VK) with the appropriate constant region primer (an equimolar mix of MHCI - MHC3 for VH and MKC for VK). Tables I & 2 detail the primers used to amplify the VH and VK region genes, respectively. In total, 12 heavy chain reactions and 11 kappa light chain reactions were performed. AmpliTaq® DNA polymerase was used to amplify the template cDNA in all cases, as follows.
[00110] The completed cDNA first strand synthesis reaction was heated at 90 °C for 5 minutes to denature the RNA-cDNA duplex and inactivate the reverse transcriptase and chilled on ice. Eleven GeneAmp™ PCR reaction tubes were labeled MKV1 -11. For each tube a 100 pi reaction mixture was prepared, each reaction mixture containing 69.3 mI of sterile water, 10 mI of 10 X PCR buffer II, 6 mI of 25 mM MgC , 2 mI each of the 10 mM stock solutions of dNTPs, 2.5 mI of 10 mM MKC primer, 2.5 mI of one of the 10 mM MKV primers and 1 mI of RNA-cDNA template mix. To each of the tubes was then added 0.7 mI of AmpliTaq® DNA polymerase and the completed reaction mix overlaid with 50 mI of mineral oil.
[00111] A similar series of reaction mixes was prepared as described above to PCR-clone the mouse heavy chain variable region gene. However, this time twelve reaction tubes were labeled and one of the twelve MHV primers and the appropriate MHC primer were added to each. That is, to PCR-amplify the variable domain gene of a mouse y1 heavy chain, for example, the MHC G1 primer was used.
[00112] The reaction tubes were loaded into a DNA thermal cycler and cycled (after an initial melt at 94 °C for 1.5 min) at 94 °C for 1 min, 50 °C for I min and 72 °C for 1 min over 25 cycles. The last cycle was followed by a final extension step at 72 °C for 10 min before cooling to 4 °C. Except for between the annealing (50 °C) and extension (72 °C) steps when an extended ramp time of 2.5 min was used, a 30 sec ramp time was used between each step of the cycle. A 10 mI aliquot from each PCR reaction was run on a 1 % (w/v) agarose /1 X TBE buffer gel containing 0.5 pg/ml ethidium bromide to determine which of the leader primers produced a PCR-product. Positive PCR-clones were about 420-500 bp in size.
[00113] The above PCR-amplification process was repeated twice more and those PCR-reactions that appeared to amplify full-length variable domain gene were selected. A 6 pi aliquot of each potential PCR-product was directly cloned into the pCR™ II vector provided by the TA Cloning® kit, as described in the manufacturers instructions. Aliquots of 10.0% (v/v), 1 .0% (v/v) and 0.1 % (v/v) aliquots of the transformed E.coli cells were pipetted onto individual 90 mm diameter LB agar plates containing 50 pg/ml ampicillin, overlaid with 25 mI of the X-Gal stock solution and 40 mI of IPTG stock solution, and incubated overnight at 37 °C. Positive colonies were identified by PCR-screening.
Table 1 - PCR primers for cloning mouse kappa light chain variable region genes
Figure imgf000034_0001
Table 2 - PCR primers for cloning mouse heavy chain variable region genes
Figure imgf000035_0001
[00114] Five mI aliquots from each PCR reaction were run on a 1 % agarose/TBE (pH 8.8) gel to determine which had produced a PCR product of the correct size (ca. 450 bp). Those putative positive PCR products so identified were directly cloned into the pCR2.1 vector provided by the TA Cloning® kit and transformed into TOP10 competent cells as described in the manufacturer's protocol. Colonies containing the plasmid with a correctly sized insert were identified by PCR-screening the colonies using the 1212 and 1233 oligonucleotide primers (Table 3) according to the method of Gilssow and Clackson (Nucleic Acids Res. 17:4000). Those putative positive clones so identified were double-stranded plasmid DNA sequenced using the ABI PRISM 310 Genetic Analyzer and the ABI PRISM BigDye™ terminator. Three positive clones each of the VH and VK genes from the B2C4 hybridoma cell line clone were sequenced, as were four positive clones of the VK gene and six of the VH gene from the B2D6 hybridoma cell line clone.
Table 3 - Primers for PCR screening and sequencing transformed colonies
Figure imgf000036_0001
[00115] The results of the 12 PCR reactions performed for each hybridoma clone (B2C4 and BCD6) to amplify the murine 11-1 F4 antibody heavy chain variable region gene are presented in Table 4(a).
[00116] The degenerate leader sequence primer MHV7, in combination with a mix of the MHCGI-3 constant region primers (Table 1 ), yielded a PCR product of about 600 bp from template cDNAs derived from both the B2C4 and B2D6 hybridoma cell lines. Since this band was larger than the expected size for an average Vhi gene (450 bp), it was not investigated further. Conversely, the degenerate leader sequence primer MHV6, in combination with a mix of the MHCGI-3 constant region primers (Table 1 ), yielded a PCR product of the expected size (450 bp) for a VH gene from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
[00117] Table 4 shows the results of the PCR amplifications performed to clone the murine 11-1 F4 monoclonal antibody variable region heavy (a) and light (b) chain genes from the SP2/0 hybridoma cell lines B2C4 and B2D6. Column three contains a record of the actual PCR results. Where a band was observed for a particular combination of primers its size in base pairs (bp) was recorded in the appropriate space.
Table 4 - Results of PCR amplification m
Figure imgf000038_0001
<b)
Figure imgf000038_0002
[00118] Sequence analysis of three clones from the B2C4 derived PCR product and five clones from the B2D6 derived PCR product revealed a single heavy chain variable region sequence (Figure 2).
[00119] The cloning strategy used (amplification of the entire variable region gene by using primers which flank this region, i.e., leader sequence and constant region sequence specific primers) allowed the complete FR1 sequence to be identified. All eight clones sequenced had identical sequence in this region (Figure 2).
[00120] The results of the 11 PCR reactions performed for each hybridoma clone (B2C4 and BCD6) to amplify the murine 11-1 F4 antibody kappa light chain variable region gene are presented in Table 4(b).
[00121] The degenerate leader sequence primer MKV6 in combination with the MKC constant region primer (Table 2), produced a PCR product of about 200 bp from template cDNA derived from the B2C4 hybridoma cell line only. Since this band was much smaller than the expected size for a VK gene (450 bp), it was not investigated further.
[00122] The degenerate leader sequence primer MKV2, in combination with the MKC constant region primer (Table 2), produced a PCR product which was smaller than the expected 450 bp band (when viewed on an agarose gel) from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines. In addition, previous VK cloning had found that the MKV2 primer amplified a well known kappa light chain pseudogene. Therefore, sequence analysis of one clone of each PCR product was performed in order to confirm that this product was a pseudogene and not the murine 11-1 F4 antibody VK gene. This sequence analysis revealed that this PCR clone was indeed the pseudogene.
[00123] Finally, the degenerate leader sequence primer MKV1 , in combination with the MKC constant region primer (Table 1 ), produced a PCR product of about the expected size (450 bp) for a VK gene, from template cDNA derived from both the B2C4 and B2D6 hybridoma cell lines.
[00124] Sequence analysis of three clones of the B2C4 derived PCR product and four clones of the B2D6 derived PCR product revealed a single kappa light chain variable region sequence which could not be identified as a pseudogene.
[00125] Thus, the 11-1 F4 antibody heavy chain variable region gene was cloned (using constant region specific and leader sequence specific primers) from the hybridoma mRNA and sequenced.
[00126] When translated, the sequence gave a TVSS peptide sequence. Analysis of 122 rearranged human VH genes, recorded in the Kabat database (Kabat et al. - Sequences of Proteins of Immunological Interest), revealed that 84% of these sequences had a TVSS peptide sequence. It was therefore concluded that the VH gene isolated was the correct 11-1 F4 antibody gene sequence.
[00127] The murine 11-1 F4 antibody variable region kappa light chain gene was also successfully cloned and sequenced, as was a non-functional VK pseudogene gene. This pseudogene was first identified by Carroll et al (Molecular Immunology (1988) 25:991 ). The sequence arises from an aberrent mRNA transcript which is present in all standard fusion partners derived from the original MOPC-21 tumor (including SP2/0). As a result of the aberrant mRNA, the invariant cysteine at position 23 is replaced by a tyrosine residue, and the VJ joint is out of frame, resulting in a stop codon at position 105.
[00128] It is common for lymphoid or hybridoma cells to synthesize more than one rearranged light immunoglobulin mRNA. These mRNAs are usually non productive due to the presence of termination codons or frame shifts not usually seen in functional VK genes. These pseudo messengers often present major problems when cloning immunoglobulin genes from hybridomas because they are very good substrates for V region PCR, despite the fact that they do not encode functional polypeptides. [00129] The 11-1 F4 antibody VK gene sequence was identified after detailed sequence analysis of seven separate PCR clones, isolated from two different PCR products to yield SEQ. ID NO: 36. Since all sequences were identical, it was accepted as the correct 11-1 F4 antibody kappa light chain variable region sequence.
[00130] The cloned VH and VK region genes were used to make the chimeric mouse-human 11-1 F4 monoclonal antibody, which was then be analyzed to confirm specific binding to AL fibrils.
EXAMPLE 2
Construction of chimeric mouse-human 11-1 F4 (c11-1 F4) antibody
[00131] In order to allow transient expression of the 11-1 F4 VH and VK variable region genes described above in mammalian cells as part of a chimeric mouse -human antibody, it was necessary to modify the 5'- and 3'- ends using specifically designed PCR primers (Table 5). The oligonucleotide primers F39836 and F39837 were used to PCR modify the 11 -1 F4 VK gene, while primers F39835 and F58933 were used to PCR modify the 11 -1 F4 VH gene. The back (BAK) primers F39836 and F39835 introduced a Hind\\\ restriction site, a Kozak translation initiation site, and an immunoglobulin leader sequence to the 5' ends of the VK and VH genes respectively. The forward (FOR) oligonucleotide primer F39837 introduced a splice donor site and a BamH\ restriction site to the 3' end of the VK gene while the forward (FOR) oligonucleotide primer F58933 appended the first 22 base pairs of the gamma-1CHi gene including an Apa\ restriction site to the 3' end of the VH gene. Table 5 - Oligonucleotide primers used to PCR modify the 11 -1 F4 heavy and kappa light chain variable region genes
Figure imgf000042_0001
[00132] The Kozak consensus sequence is crucial to the efficient translation of a variable region sequence (Kozak - J Mol Bio 196:947). It defines the correct AUG codon from which a ribosome commences translation, and the single most critical base is the adenine (or less preferably, a guanine) at position -3, upstream of the AUG start.
[00133] The immunoglobulin leader sequence ensures that the expressed antibody is secreted into the medium and therefore is easily harvested and purified. The leader sequences used in this instance were the murine 1 1 -1 F4 \A< and VH leader sequences cloned from the hybridoma cDNA during the VH and VK cloning process.
[00134] The splice donor sequence is important for the correct in-frame attachment of the light chain variable region to its appropriate constant region, thus splicing out the 130 bp VK:CK intron. The heavy chain variable region was attached directly to its appropriate constant region gene via the Apa\ site, thus eliminating the need for a splice donor site.
[00135] The sub-cloning restriction sites Hind III and SamHI, and Hind III and Apal, respectively, bracket the modified Vi< and VH variable region genes, while the use of different unique restriction sites ensured directional sub- cloning into the appropriate mammalian expression vector. [00136] The 11-1 F4 light chain variable region gene was first carefully analyzed to identify any unwanted splice donor sites, splice acceptor sites, and Kozak sequences (see Table 6). Both the heavy and light chain variable region genes were analyzed for the presence of any extra sub-cloning restriction sites which would later interfere with the subcloning and/or expression of functional whole antibody. None were found.
Table 6 - Sequences important for the efficient expression of
immunoglobulin genes in mammalian cells
Figure imgf000043_0001
[00137] Separate PCR reactions were prepared as follows, one for each variable region gene. The plasmids 11-1 F4 VH.PCR2.1 and 11 -1 F4 VK.PCR2.1 described above were used as templates. A 100 pi reaction mixture was prepared in each PCR tube, each mixture containing up to 41 pi of sterile water, 10 mI of 10 x PCR buffer I, 8 mI of the 10 mM stock solution of dNTPs, 1 m I of 10 mM of 5' forward primer, 1 mI of the 10 mM 3' Reverse primer, and 1 m I of a 1/10 dilution of template DNA. Finally, 0.5 mI of AmpliTaq® DNA polymerase (2.5 units) was added before overlaying the completed reaction mixture with 50 mI of mineral oil. The reaction tubes were loaded into a DNA thermal cycler and cycled (after an initial melt at 94 °C for 1 min) at 94 °C for 30 sec, 68 °C for 30 sec and 72 °C for 50 sec over 25 cycles. The completion of the last cycle was followed with a final extension step at 72 °C for 7 min before cooling to 4 °C. A 10 m I aliquot from each PCR reaction tube was run on a 1.2% (w/v) agarose/ 1 X TBE buffer gel containing 0.5 pg/ml ethidium bromide to determine size and presence of a PCR- product. Positive PCR- clones were about 420bp in size. Those putative positive PCR products so identified were directly cloned into the pCR2.1 vector, provided by the Topo TA Cloning® kit, and transformed into TOP10 competent cells as described in the manufacturer's protocol. Colonies containing the plasmid with a correctly sized insert were identified by PCR-screening the colonies using the 1212 and 1233 oligonucleotide primers (Table 3) according to the method of Gilssow and Clackson. Those putative positive clones so identified were double-stranded plasmid DNA sequenced using the ABI PRISM 310 Genetic Analyzer and the ABI PRISM BigDye™ terminator. Two positive clones each of the Topo TA cloned Vhi and VK genes were sequenced.
[00138] Clones containing the correctly modified 11-1 F4 VH and 11-1 F4 VK genes were identified and the modified V genes from these clones were subcloned into their respective expression vectors to facilitate the expression of chimeric heavy and kappa light chains in mammalian cells. The modified 11-1 F4 VK gene was subcloned into the expression vector pKN100 (Figure 4) as a Hind\\\-BamH\ fragment; this vector contains a human kappa constant region gene (allotype: Km (3 Ala153, Ser191)). The modified 11-1 F4 VH gene was also subcloned as a Hind\\\-Apa\ fragment into the expression vector pG1 D200 (Figure 5); this vector contained a human y1 constant region gene (allotype: G1 m (-1 Glu377, Met38l, -2 Ala462, 3 Arg222, Ser229)). Both the kappa and y1 constant region allotypes used are commonly found in the Caucasian population. The ligated expression constructs, 11-1 F4VK.pKN100 and 11- 1 F4VH.pG1 D200, were then used to transform DH5ct competent cells, and positive clones were identified using the PCR screening method discussed above with the original PCR modification primers (Table 4). The expression vectors are readily available. EXAMPLE 3
Construction of a single supervector for transient expression of chimeric 11-1 F4 in COS Cells.
[00139] A single supervector expressing both immunoglobulin chains of the chimeric 11-1 F4 antibody was constructed as follows. The 11-1 F4 kappa light chain expression cassette (which contained the HCMVi promoter, the 11- 1 F4 kappa light chain variable region gene, and the kappa light chain constant region gene) was restriction enzyme digested (EcoRI at positions 1 and 2490) out of the 11-1 F4VK.pKN100 construct (Figure 4) and subsequently ligated into the 11-1 F4VHpG1 D200 construct via the unique EcoRI (position 4297, Figure 5). This ligation resulted in the construction of a supervector construct, pG1 KD200-11-1 F4, containing both the heavy and kappa light chains of the 11 -1 F4 chimeric antibody.
EXAMPLE 4
Transient expression of the chimeric V1/K.11-IF4 whole antibody in COS cells
[00140] The chimeric 11-IF4 antibody was transiently expressed in COS cells from the European Collection of Cell Cultures (ECACC) in two ways:
(i) By cotransfection of 10pg of each of the vector constructs 11- 1 F4VK.pKN100 and 11-1 F4VFI.pG1 D200. Co-transfections were carried out in duplicate.
(ii) By transfection of I3pg of the single supervector construct pG1 KD200-11 -1 F4. Supervector transfections were carried out five times.
[00141] The following transfection method was used. The COS cell line was grown in DMEM supplemented with 10% (v/v) FCS, 580 pg/ml L- glutamine and 50 Units/ml penicillin/ 50 pg/ml streptomycin (“media”) in a 150 cm2 flask until confluent. The cells were trypsinized, spun down in a bench top centrifuge (250 g for 5 min), then re-suspended in 6 ml of media before dividing them equally between three 150 cm2 flasks, each containing 25 ml of fresh, pre-warmed media. The cells were incubated overnight at 37 °C in 5% CO2 and then harvested the next day while they are still growing exponentially. Each flask contained approximately 1 x 107 cells. The cells were trypsinized again, pelleted as before, and washed in 20 ml of PBS, following which they were re-suspend in sufficient PBS to create a cell concentration of 1 x I07 cells/ml. 700 pi of these washed COS cells were pipetted into a Gene Pulser® cuvette, to which was then added 1 mI of both the heavy chain and kappa light chain expression vector DNA (each at 10 pg) or 13 pg of the super-vector construct. A 1900 Volt, 25 pFarad capacitance pulse was delivered to the mixture using the Bio-Rad Gene Pulser® apparatus. The pulsing was repeated for each experimental transfection and a "no DNA" control (in which the COS cells were electroporated in the absence of any DNA). A positive control of a previously-expressed antibody was also carried out to test the efficiency of the COS cells.
[00142] The COS cells were allowed to recover at room temperature for 10 min, then gently pipetted the into a 10 cm diameter tissue culture dish containing 8 ml of pre- warmed DMEM supplemented with 10% (v/v) y - globulin free FBS, 580 pg/ml L- glutamine and 50 Units/ml penicillin / 50 pg/ml streptomycin, and incubated in 5% CO2 at 37 °C for 72 hours before harvesting the COS cell supernatant for analysis. After incubation for 72 hours the medium was collected, spun to remove cell debris and analyzed by ELISA for chimeric antibody production and antigen binding of the c11-1 F4 antibody.
EXAMPLE 5
Quantification of the chimeric V1 /K 1 1 -1 F4 antibody via capture ELISA
[00143] Following expression, the whole IgG molecules present in the COS cell supernatant were quantified using a capture ELISA assay. IgG molecules were captured on a Nunc-lmmuno MaxiSorb™ plate via an immobilized goat anti-human IgG, Fey fragment - specific antibody, and detected via an anti-human kappa light chain peroxidase conjugated antibody. A standard curve was generated by capturing and detecting known concentrations of a standard IgG antibody on the same plate in the same way as follows. Each well of a 96-well immunoplate was coated with 100 mI aliquots of 0.4 pg/ml goat anti-human IgG antibody diluted in PBS and incubated overnight at 4 °C. The excess coating solution was removed and the plate was washed three times with 200 m l/well of washing buffer (1xPBS, 0.1 % TWEEN). Into all wells except the wells in column 2, rows B to G, was dispensed 100mI of SEC buffer. A 1 pg/ml solution of the human lgG1/kappa antibody in SEC buffer was prepared to serve as a standard and 200 m l/well was pipetted into the wells in column 2, rows B and C. The medium from the transfected cos cells was centrifuged (250g, 5 min), saving the supernatant. An aliquot of 200 mI of the supernatant from the "no DNA" control (in which COS cells were transfected in the absence of DNA) was pipetted into the well in column 2, row D, and aliquots of 200 m l/well of experimental supernatants were pipetted into the wells in column 2, rows E, F, and G. The 200 mI aliquots in the wells of column 2, rows B to G were mixed and then 100 mI was transferred from each well to the neighboring well in column 3. This process was continued to column 11 with a series of 2-fold dilutions of the standard, control, and experimental samples, following which all were incubated at 37 °C for 1 hour and all the wells were rinsed six times with 200 mI aliquots of washing buffer. The goat anti-human kappa light chain peroxidase conjugate was diluted 5000-fold in SEC buffer and 100 mI of the diluted conjugate added to each well, followed by a repetition of the incubation and rinsing steps. To each well was added 150mI of K-BLUE substrate, followed by incubation in the dark at 25 °C for 10min. The reaction was stopped by adding 50 mI of RED STOP solution to each well and the optical density was read at 655nm.
EXAMPLE 6
Binding analysis of the chimeric 11-1 F4 antibody
[00144] The chimeric 11-1 F4 antibody was tested for binding to amyloid fibrils using a direct binding ELISA assay. Synthetic fibrils were formed from an immunoglobulin light chain protein and used to monitor the reactivity of the antibody in a solid-state ELISA-based assay using a "low- binding" polystyrene plates (Costar,# 3474). Immediately prior to coating the plate, a mass of 250 pg of fibrils was diluted to 1 ml with coating buffer (0.1 % bovine serum albumin in phosphate buffered saline pH 7.5). The sample was then sonicated for 20 sec using a Tekmar Sonic Disruptor sonicating probe, with the power set to 40% of maximum, resulting in a solution of short fibrils composed of up to 2-5 protofiliments each. This solution was then diluted to 5 ml, mixed well by vortex, and aliquoted into the wells of the plate. This process yielded 50 pi of fibril solution having a concentration of 50 pg/ ml in each well. The plate was then dried overnight by placing it uncovered in a 37 °C incubator.
[00145] The ELISA assay was then performed as follows within 48 hours of preparing the plate. The wells were blocked by the addition of 100 pi of l% BSA in PBS and incubated for 1 hour at room temperature on a shaker. The plate was washed x3 in PBS, 0.05% Tween 20 (v/v). To each well of the plate was added 50 pi of a solution of c11-1 F4 (3pg/ml antibody in 0.1% BSA/PBS) and the plate incubated at room temperature for 1 hour on a shaker. The plate was again washed x3 (as before) and detection of bound antibody was accomplished using a biotinylated goat anti-mouse IgG antibody (Sigma# B- 8774, anti-heavy and light-chain).
[00146] Sequence analysis of the successfully modified VH and VK genes revealed the correct sequence was present. Detailed DNA and amino acid sequences of the modified 11 -1 F4 VK and VH genes are presented in Figures 3 & 4. The modified VK and VH genes were successfully cloned into the mammalian expression vectors pG1 D200 and pKN100 respectively, and the resulting 11-1 F4VK.pKN100 and 11-1 F4VHpG1 D200 constructs were used for cotransfection of mammalian cells.
[00147] The 11-1 F4VK.pKN100 and 11-1 F4VHpG1 D200 constructs were also subsequently used to construct a single supervector (pG1 KD200-11-1 F4), which expressed the chimeric 11 - 1 F4 antibody in mammalian cells. The chimeric 11-1 F4 antibody expression levels, from both cotransfections and supervector transfections of ECACC COS cells were assayed. The expression levels observed from the pG1 KD200-11-1 F4 supervector transfections (10326 ng/ml) were 3.7 fold higher than the levels observed from the corresponding co-transfections of the 11 -1 F4VK.pKN100 and 11- 1 F4VFIpG1 D200 constructs (2820 ng/ml).
[00148] Following expression and quantification, the chimeric 11-1 F4 antibody was tested for binding to target antigen (amyloid fibrils kindly supplied by the NCI) by direct binding ELISA. The results of the binding ELISA are presented in Figure 8. Supenatants from the two best individual pG1 KD200- 11-1 F4 supervector transfections were assayed in parallel with one supernatant from the corresponding co-transfection.
[00149] The results indicated that the chimeric 11-1 F4 antibody bound to the amyloid fibrils with a higher affinity than its murine equivalent. This result is surprising and unexpected because normally a chimeric antibody would be expected to have a binding affinity comparable to the original murine antibody. Without intending to be bound by the particular mechanism, the inventors believe it is possible that the net effect of combining the 11-1 F4 murine V regions with the human g1/k C regions used to create the chimeric 11-1 F4 antibody produced an antibody of higher affinity.
[00150] Samples of CHO cells (identified as CAEL-101 ) that secrete one embodiment of the chimeric 11-1 F4 monoclonal antibody (which embodiment is sometimes referred to herein as CAEL-101 or as the antibody CAEL-101 ) used herein were deposited with the American Type Culture Collection (ATCC Acc. No: PTA-125146) on June 27, 2018, in compliance with the Budapest Treaty.
EXAMPLE 7
Imaging Fluman Amyloid Deposits in Mice
[00151] Human amyloid extracts from the heart (K1 ), liver (K1 ), spleen (A1 ) and kidney (A6) were kindly provided by Tufts University. Lyophilized human amyloid extracts were suspended in 25ml of sterile PBS and homogenized for 3 minutes and centrifuged at 12,000g for 30 minutes. 100mg of the resulting pellet was resuspended in sterile saline. Balb/c mice were then injected subcutaneously with amyloid extract.
P24l 1CAEL-101 :
[00152] cGMP grade CAEL-101 was radiolabeled with 124l, a positron emitting radioisotope used for PET imaging, with the standard iodegen reaction. (Fraker, et al. - Biochem Biophys Res Commun 80(4): 849-57; Markwell et at. - Biochem 17:4807-17) Approximately 5 days after human amyloid extract was implanted to form subcutaneous amyloidomas, the mice were injected with 50-200pCi of [124l ]CAEL-101 and imaged 1 , 4 and 7 days post injection using an Inveon microPET scanner. SUVmax for amyloidomas and contralateral background were obtained by drawing regions of interest in the PMOD software package and calculating tumor-to-background (amyloid deposit-to-background) (T:B) ratios at 1 and 4 days post tracer infusion.
[00153] [124I]CAEL-101 successfully imaged 100% of mice bearing human amyloid extracts (k1 , A1 and A6 subtypes derived from heart, liver, spleen, and kidney). We demonstrated for the first time imaging of cardiac derived amyloidosis (shown in Figure 9), which was reported to not work in prior literature. (Wall, et at. - Blood. 2010 116:2241 ). Human amyloidomas were visualized at both 1 and 4 days post tracer infusion, with significantly increasing T:B ratio by day 4. T:B ratios ranged from 2.1 to 4.2 at 4 days. We found heterogeneous uptake among various amyloidomas. For example, mice implanted with k subtypes demonstrated significantly better in vivo T:B ratios (4.1 +/- 0.20), compared to l subtypes (2.8 +/- 0.46), although all amyloidomas exhibited T:B uptake > 2.1 , which would be clinically significant.
[00154] This result of heterogeneous uptake among the various amyloidomas supports using real time PET imaging to stratify patients for therapy with the chimeric or humanized antibody described herein as well as to determine the appropriate dose of the antibody therapy. That is, one may determine the dosing of a patient based on the detected uptake of the labeled antibody or antibody fragment. A patient showing strong uptake (affinity) for the labeled antibody or antibody fragment may require a smaller amount of therapeutic antibody or antibody fragment than a patient showing a weaker uptake (affinity) for the labeled antibody or antibody fragment. The present disclosure therefore provides a method of determining the appropriate dosage of the disclosed humanized or chimeric antibody for therapy comprising the steps of administering to the patient a labeled antibody or antibody fragment disclosed herein, determining the uptake of the labeled antibody or antibody fragment by the amyloid deposits of the patient, and administering a dose or series of doses of chimeric or humanized antibody or antibody fragment to the patient based on the determined amount of uptake of the labeled antibody or antibody fragment.
f89Zr 1CAEL-101 :
[00155] Although [124l ]CAEL-101 demonstrated significant binding to amyloidamas, the 124l radiolabel dehalogenates, causing much of the detected activity to be in the thyroid or untargeted (if thyroid is blocked). We therefore sought a method to radiolabel with a more stable radioisotope that can be imaged after blood clearance of unbound radiolabel (7-10 days). We radiolabeled CAEL-101 with 89-Zirconium (89Zr), using two different strategies to create [89Zr] -CAEL-101. One strategy employed the standard NCS linker that binds to random lysines on an antibody. ( Eur J Nucl Mol Imaging. 2010 Feb. 37(2): 250-59; Curr Radiopharm. 2011 Apr 1 ; 4(2): 131-139) This method is used in many clinical applications but has been shown to create multiple radioisomers that can theoretically alter binding or biodistribution between batches. We therefore also radiolabeled CAEL-101 using a site specific approach that targets cystienes using the bifunctional deferoxamine- maleimide cross linker. (Nuclear Medicine and Biology 37 (2010) 289-297) Approximately 5 days after human cardiac amyloid extract was implanted to form subcutaneous amyloidomas, animals were injected with 50-200pCi of [89Zr]CAEL-101 and imaged 1 , 4, 7, 1 1 and 14 days post injection using an Inveon microPET scanner.
[00156] Both variants of [89Zr]CAEL-101 successfully imaged mice bearing cardiac amyloid extracts. Human amyloidomas were visualized very well throughout the entire term of the experiment (14 days post tracer injection) with no evidence of significant breakdown of radiotracer. This is in contrast to what we found with the [124I]CAEL-101 that had significantly broken down by day 4 and 7. This long duration of imaging of amyloid deposits enables one to use this labeled antibody for imaging deposits after significant blood clearance of unbound labeled antibody, which improves tumor-to- background (amyloid deposit-to-background) ratios and is important for imaging disease in organs that have high adjacent blood compartments, such as cardiac amyloid deposition. We found long term in vivo stability of the antibody binding component as we observed amyloidoma even 14 days after tracer injection. We again demonstrated imaging of cardiac derived amyloidosis, which had been reported to not work in prior literature. We observed for the first time ipsilateral uptake in lymphatic tissue, likely caused by amyloid in immune cells after phagocytosis migrating to draining nodes, possibly due to the fact that we implanted the amyloidomas in immunocompetent mice. This model undergoes spontaneous amyloid clearance due to the presence of human amyloid. This novel observation may provide a basis for a method of imaging response to therapy by looking for PET tracer uptake in lymph nodes.
[00157] In the description and claims of this specification the word “comprise” and variations of that word, such as“comprises” and“comprising” are not intended to exclude other features, additives, components, integers or steps but rather, unless otherwise stated explicitly, the scope of these words should be construed broadly such that they have an inclusive meaning rather than an exclusive one. [00158] Although the compositions and methods of the invention have been described in the present disclosure by way of illustrative examples, it is to be understood that the invention is not limited thereto and that variations can be made as known by those skilled in the art without departing from the teachings of the invention defined by the appended claims.

Claims

CLAIMS What is claimed is:
1. An in vivo method of detecting the presence, location and/or quantity of amyloid deposits in a patient suspected of having amyloid deposits which comprises administering to the patient an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising:
a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51 ; and
detecting the presence, location, and/or quantity of amyloid deposits by detecting the detectable label bound to the amyloid deposits by diagnostic imaging.
2. The method of claim 1 wherein the antibody or antigen binding fragment comprises a chimeric mouse-human antibody which comprises a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
3. The method of claim 1 wherein the method of detection is positron emission spectroscopy (PET).
4. The method of claim 3, wherein the detectable label is 124l.
5. The method of claim 3, wherein the detectable label is 89Zr.
6. The method of claim 4 wherein the antibody is chimeric 11 -1 F4.
The method of claim 5 wherein the antibody is chimeric 11 -1 F4.
7. The method of claim 4 wherein the antibody is CAEL-101.
8. The method of claim 5 wherein the antibody is CAEL-101.
9. The method of claim 1 , wherein the amyloid deposits are in the heart.
10. A composition for detecting the presence, location, and/or quantity of amyloid deposits in a subject which comprises an antibody or antigen binding fragment having a detectable molecule linked thereto, the antibody or antigen-binding fragment comprising:
a variable heavy chain (VH) comprising: a complementarity determining region (CDR) H1 comprising SEQ ID NO: 53; a CDRH2 comprising SEQ ID NO: 53; and a CDRH3 comprising SEQ ID NO: 54; and
a variable light chain (VK) comprising a CDRL1 comprising SEQ ID NO: 49; a CDRL2 comprising SEQ ID NO: 50; and a CDRL3 comprising SEQ ID NO: 51.
11. The composition of claim 10, wherein the antibody or antigen binding fragment comprises a chimeric mouse-human antibody which comprises a VK region comprising SEQ ID NO: 47 and a VH region comprising SEQ ID NO: 48.
12. The composition of claim 11 , wherein the detectable molecule is 124l.
13. The composition of claim 11 , wherein the detectable molecule is 89Zr.
14. The composition of claim 12 wherein the antibody is chimeric 11 -1 F4.
15. The composition of claim 13 wherein the antibody is chimeric 11 -1 F4.
16. The composition of claim 12 wherein the antibody is CAEL-101.
17. The composition of claim 13 wherein the antibody is CAEL-101.
18. The composition of claim 11 wherein the amyloid deposits are in the heart.
19. A method of monitoring disease progression in a patient diagnosed with an amyloid deposition disease and having amyloid deposits comprising the steps of: a. administering to said patient the composition of claim 10 and conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, b. treating the patient with a therapy intended to remove amyloid deposits,
c. administering to said patient the composition of claim 10, d. conducting diagnostic imaging on the patient to detect the amount of detectable molecule bound to the amyloid deposits, and
e. comparing the detected amount of detectable molecule in step d to the detected amount of detectable molecule in step a.
20. The method of claim 18, wherein the patient is treated with humanized or chimeric 11 -1 F4 antibody or antigen-binding fragment thereof
21. The method of claim 19 wherein the composition is the composition of claim 11.
22. The method of claim 20, wherein the method of detection is PET.
23. The method of claim 21 wherein the detectable label is 124l.
24. The method of claim 21 , wherein the detectable label is 89Zr.
25. The method of claim 22 wherein the antibody is chimeric 11 -1 F4.
26. The method of claim 23 wherein the antibody is chimeric 11 -1 F4.
27. The method of claim 22 wherein the antibody is CAEL-101.
28. The method of claim 23 wherein the antibody is CAEL-101.
29. The method of claim 23 wherein the amyloid deposits are in the heart.
30. A method of determining the effectiveness of treatment to remove amyloid deposits in a patient comprising a) treating the patient with a therapeutically-effective dose of the chimeric 11-F4 antibody or antigen- binding fragment thereof, b) administering to the patient a diagnostically-effective amount of the diagnostic composition of claim 10, and c) measuring by diagnostic imaging the amount of detectable molecule from the diagnostic composition in the lymph nodes of the patient, wherein the higher the amount of detectable molecule detected in the lymph nodes, the more effective the treatment.
31. The method of claim 31 , wherein the amyloid deposits are in the heart.
PCT/US2019/058720 2018-10-31 2019-10-30 Methods and compositions for imaging amyloid deposits WO2020092474A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2021523841A JP7337922B2 (en) 2018-10-31 2019-10-30 Methods and compositions for imaging amyloid deposits
EP19880738.0A EP3873932A4 (en) 2018-10-31 2019-10-30 Methods and compositions for imaging amyloid deposits

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862753410P 2018-10-31 2018-10-31
US62/753,410 2018-10-31

Publications (1)

Publication Number Publication Date
WO2020092474A1 true WO2020092474A1 (en) 2020-05-07

Family

ID=70462389

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/058720 WO2020092474A1 (en) 2018-10-31 2019-10-30 Methods and compositions for imaging amyloid deposits

Country Status (3)

Country Link
EP (1) EP3873932A4 (en)
JP (2) JP7337922B2 (en)
WO (1) WO2020092474A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100080806A1 (en) * 2004-08-19 2010-04-01 Yanxin Liu Anti-human trail receptor dr5 monoclonal antibody (ad5-10), method thereof and use of the same
US20180148504A1 (en) * 2016-11-23 2018-05-31 Morphotek, Inc. Anti-Folate Receptor Alpha Antibodies And Uses Thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090297439A1 (en) 2008-06-02 2009-12-03 Metheresis Translational Research Sa, Anti-met monoclonal antibody, fragments and derivatives thereof for use in tumor diagnosis, corresponding compositions and kits
US10213506B2 (en) 2014-08-26 2019-02-26 University Of Tennessee Research Foundation Targeting immunotherapy for amyloidosis
KR102607629B1 (en) 2017-06-29 2023-12-01 더 트러스티스 오브 콜롬비아 유니버시티 인 더 시티 오브 뉴욕 Chimeric antibodies for the treatment of amyloid deposition diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100080806A1 (en) * 2004-08-19 2010-04-01 Yanxin Liu Anti-human trail receptor dr5 monoclonal antibody (ad5-10), method thereof and use of the same
US20180148504A1 (en) * 2016-11-23 2018-05-31 Morphotek, Inc. Anti-Folate Receptor Alpha Antibodies And Uses Thereof

Also Published As

Publication number Publication date
JP2023093673A (en) 2023-07-04
JP2022512892A (en) 2022-02-07
EP3873932A4 (en) 2022-11-23
EP3873932A1 (en) 2021-09-08
JP7337922B2 (en) 2023-09-04

Similar Documents

Publication Publication Date Title
AU2018311688B2 (en) Methods and compositions for treatment of amyloid deposition diseases
US6329507B1 (en) Dimer and multimer forms of single chain polypeptides
FI117509B (en) Humanized antibodies to the leukocyte adhesion molecule VLA-4
EA013677B1 (en) Human monoclonal antibodies against cd25 and use thereof
MX2010011955A (en) Dual variable domain immunoglobulins and uses thereof.
US20230212273A1 (en) Chimeric antibodies for treatment of amyloid deposition diseases
CA2837472A1 (en) Rationally-designed anti-mullerian inhibiting substance type ii receptor antibodies
US20200002410A1 (en) Methods and compositions for imaging amyloid deposits
US20240124595A1 (en) Antibodies to igf2r and methods
JP7337922B2 (en) Methods and compositions for imaging amyloid deposits
CN111108120A (en) Antibody against human vascular endothelial growth factor receptor and application thereof
US20230391873A1 (en) Methods for treating mismatch repair deficient locally advanced rectal cancer using dostarlimab
JP5990523B2 (en) Imaging diagnostic agent containing anti-human transferrin receptor antibody
Broisat et al. Nanobodies Targeting Mouse/Human VCAM1 for the Nuclear Imaging of Atherosclerotic Lesions First author’s surname and short title
TW201305211A (en) Novel homogeneous humanized antiproliferation antibodies

Legal Events

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

Ref document number: 19880738

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021523841

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019880738

Country of ref document: EP

Effective date: 20210531