WO2023105051A1 - Cancer therapy targeting egfr - Google Patents

Cancer therapy targeting egfr Download PDF

Info

Publication number
WO2023105051A1
WO2023105051A1 PCT/EP2022/085198 EP2022085198W WO2023105051A1 WO 2023105051 A1 WO2023105051 A1 WO 2023105051A1 EP 2022085198 W EP2022085198 W EP 2022085198W WO 2023105051 A1 WO2023105051 A1 WO 2023105051A1
Authority
WO
WIPO (PCT)
Prior art keywords
egfr
component
seq
nos
amino acid
Prior art date
Application number
PCT/EP2022/085198
Other languages
French (fr)
Inventor
Athanasios PALLIS
Original Assignee
Les Laboratoires Servier
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 Les Laboratoires Servier filed Critical Les Laboratoires Servier
Publication of WO2023105051A1 publication Critical patent/WO2023105051A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • Cancer ranks as a leading cause of death and an important barrier to increasing life expectancy in every country of the world. According to estimates from the World Health Organization (WHO) in 2019, cancer is the first or second leading cause of death before the age of 70 years in 112 of 183 countries and ranks third or fourth in a further 23 countries.
  • WHO World Health Organization
  • Colorectal cancer is the third most common cancer type in the world in terms of incidence and the second one in terms of mortality, with an estimated 1 ,800,000 new cases and 881 ,000 deaths in 2018.
  • mCRC metastatic colorectal cancer
  • Anti-PD-1 agents such as nivolumab and pembrolizumab have seen initial success in microsatellite instability high/defective mismatch repair (MSI-H/dMMR) mCRCs, although these comprise only 1 % of patients.
  • Tumors that are RAS/BRAF wild-type (WT) and EGFR-positive are among the most common gene expression profiles, accounting for approximately 40% of CRCs.
  • EGFR Epidermal Growth Factor Receptor
  • the present invention is based on therapies for enhancing immunity comprising one or two anti-EGFR antibodies, e.g., as described herein, with trifluridine/tipiracil composition.
  • the therapy is for treating cancer.
  • pharmaceutical compositions comprising the components of the therapies, and use of the therapies for enhancing immunity (e.g., treating cancer) in a patient.
  • the therapies described herein may be used in a method for enhancing immunity (e.g., treating cancer) in a patient; may be used for the manufacture of a medicament for enhancing immunity (e.g., treating cancer) in a patient; or may be for use in enhancing immunity (e.g., treating cancer) in a patient.
  • enhancing immunity it is understood at least the blockage of the interaction between the ligandbinding domain of EGFR with EGFR ligands and concomitantly, the resultant intracellular signaling pathway.
  • EGFR expression is frequently accompanied by the production of EGFR-ligands, TGF-alpha and EGF among others, by EGFR-expressing tumour cells which suggests that an autocrine loop participates in the progression of these cells (Baselga, etal.(1994) Pharmac. Therapeut. 64: 127-154; Modjtahedi, et al. (1994) Int. J. Oncology. 4: 277-296).
  • the present disclosure provides a combination comprising : a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof that competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component.
  • the heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 of the anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprise the amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively.
  • the heavy chain variable domain (VH) and light chain variable domain (VL) of the anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
  • the heavy chain (HC) and light chain (LC) of the anti-EGFR component comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
  • the heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 of an anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 1 -6, and the H-CDR1-3 and L-CDR1 -3 of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 11 -16.
  • the heavy chain variable domain (VH) and light chain variable domain (VL) of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 7 and 8
  • the VH and VL of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 17 and 18.
  • the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
  • HC heavy chain
  • LC light chain
  • the method of enhancing immunity in a human patient in need thereof comprises administering to the patient : a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof that competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component.
  • the anti-EGFR component comprises an anti- EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 that comprise the amino acid sequences of (a) SEQ ID NOs: 1 -6, respectively; or (b) SEQ ID NOs: 11 -16, respectively.
  • the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of (a) SEQ ID NOs: 7 and 8, respectively; or (b) SEQ ID NOs: 17 and 18, respectively.
  • the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of (a) SEQ ID NOs: 9 and 10, respectively; or (b) SEQ ID NOs: 19 and 20, respectively.
  • HC heavy chain
  • LC light chain
  • the method comprises administering the anti- EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H- CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 that comprise the amino acid sequences of SEQ ID NOs: 1 -6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with H- CDR1 -3 and L-CDR1 -3 that comprise the amino acid sequences of SEQ ID NOs: 11 -16, respectively.
  • H- CDR heavy chain complementarity-determining regions
  • L-CDR light chain complementarity-determining regions
  • the anti-EGFR component comprises an anti- EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti- EGFR antibody or an antigen-binding portion thereof with a VH and VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • the anti-EGFR component comprises an anti- EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
  • the anti-EGFR component may be for example futuximab, modotuximab, or futuximab + modotuximab.
  • the method comprises administering to the patient: a) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 1-6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 11 -16, respectively; and a trifluridine/tipiracil component ; b) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 17 and 18, respectively; and a trifluridine/tipiracil component ; or c) an anti-EGFR component comprising an anti
  • the one or two anti-EGFR antibodies or antigen-binding portions thereof and the trifluridine/tipiracil component may be administered to the patient concurrently or sequentially.
  • the patient has cancer, e.g., a hematological malignancy or a solid tumor.
  • the cancer is colorectal cancer or gastric cancer.
  • the patient has a metastatic colorectal cancer (mCRC).
  • the anti-EGFR component is administered at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, (e.g. weekly).
  • the trifluridine/tipiracil component is administered at a dose of 20 to 80 mg/m 2 /day, optionally the component trifluridine/tipiracil is divided in two portions per day for five consecutive days and then a 2-days rest period.
  • the trifluridine/tipiracil component is administered at a dose of 20mg/m 2 , 25mg/m 2 , 30mg/m 2 , 35mg/m 2 .
  • the one or two anti-EGFR antibodies or antigen-binding portions thereof are formulated for intravenous infusion.
  • the trifluridine/tipiracil component is formulated for orally administration.
  • the present disclosure provides a method of treating an advanced solid tumor malignancy in a patient, comprising administering to the patient an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and a trifluridine/tipiracil component, wherein the anti-EGFR component is administered by IV infusion at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, wherein the trifluridine/tipiracil component is administered orally at a dose of 35mg/m 2 .
  • the present disclosure provides a method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; for example, the cancer is locally advanced and/or unresectable, and for example the patient has failed on first-line standard of care therapy and/or second-line standard of care therapy.
  • the anti-EGFR antibody component is administered at a loading dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , and a maintenance dose of 6mg/kg day 8, day 15, day 22 of Cycle 1 each cycle and days 1 , 8 , 15, 22 of all other 28-days cycles, and the trifluridine/tipiracil component is administered at a dose of 35mg/m 2 on days 1 to day 5 and days 8 to day 12 of each 28 days cycle, wherein the anti- EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally.
  • the present disclosure provides a method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; wherein the patient is (i) without RAS mutations in any of the following codons: codons 12 and 13 in exon 2, codons 59 and 61 in exon 3, and codons 117 and 146 in exon 4; and/or (ii) without the BRAF V600E mutation.
  • the anti-EGFR antibody component is administered at a loading dose dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , day 8, day 15, day 22 of each cycle, and the trifluridine/tipiracil component is administered at a dose of 35mg/m 2 on days 1 to day 5 and days 8 to day 12 of each cycle, wherein the anti-EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally.
  • C1 D1 Cycle 1 Day 1
  • the trifluridine/tipiracil component is administered at a dose of 35mg/m 2 on days 1 to day 5 and days 8 to day 12 of each cycle, wherein the anti-EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally.
  • a method of the present disclosure further comprises administering to the patient radiation therapy, or at least one of a chemotherapeutic agent, an anti-neoplastic agent, and an anti-angiogenic agent.
  • Treatment according to a method of the present disclosure may result in tumor regression, delay of tumor progression, inhibition of cancer progression, inhibition of cancer metastasis, prevention of cancer recurrence or residual disease, and/or prolonged survival.
  • treatment according to a method of the present disclosure may result in an improved objective response rate, improved clinical benefit rate, improved duration of response, increased progression-free survival, increased overall survival, or any combination thereof, e.g., in comparison to an untreated patient.
  • the present disclosure also provides a pharmaceutical composition comprising an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as described herein, and further comprising a trifluridine/tipiracil component, and a pharmaceutically acceptable excipient.
  • the present disclosure also provides a pharmaceutical composition comprising the components of the method as described herein.
  • the pharmaceutical composition may comprise the antibodies or antigen-binding portions of any of the methods described herein, and may be for use in treating a human patient in any of the methods described herein.
  • an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein for use in enhancing immunity in a human patient in need thereof in combination with a trifluridine/tipiracil component.
  • an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for use in treating a human patient in a method as described herein.
  • the present disclosure also provides use of an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for the manufacture of a medicament for enhancing immunity in a human patient in need thereof.
  • the present disclosure also provides use of an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for the manufacture of a medicament for treating a human patient in a method as described herein.
  • FIG. 1 study design
  • FIG.2 study plan for the Safety Lead-In Part
  • FIG.3 study plan for the Phase 3 Part
  • the present disclosure provides new combination therapies and compositions that target human EGFR by using antibodies and trifluridine/tipiracil composition.
  • the therapies i.e., combination therapies
  • compositions can be used to treat cancer in a human patient.
  • EGFR refers to the human forms of this target.
  • a human EGFR polypeptide sequence is available under UniProt Accession No. P00533 (SEQ ID NO: 21 ).
  • antibody refers to a tetramer comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa) inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable domain (VH) and a heavy chain constant region (CH).
  • Each light chain is composed of a light chain variable domain (VL) and a light chain constant region (CL).
  • VH and VL domains can be subdivided further into regions of hypervariability, termed “complementarity determining regions” (CDRs), interspersed with regions that are more conserved, termed “framework regions” (FRs).
  • CDRs complementarity determining regions
  • FRs frame regions
  • Each VH and VL is composed of three CDRs (H-CDR herein designates a CDR from the heavy chain; and L-CDR herein designates a CDR from the light chain) and four FRs, arranged from amino-terminus to carboxyl-term inus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • the assignment of amino acid numbers, and of FR and CDR regions, in the heavy or light chain may be in accordance with IMGT® definitions (Lefranc et al., Dev Comp Immunol (2003) 27(1):55-77); Eu numbering; or the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and 1991 )); Chothia & Lesk, J. Mol. Biol. (1987) 196:901-17; Chothia et al., Nature (1989) 342:878-83; MacCallum et al., J. Mol. Biol. (1996) 262:732-45; or Honegger and Pluckthun, J. Mol. Biol. (2001 ) 309(3):657-70.
  • recombinant antibody refers to an antibody that is expressed from a cell or cell line comprising the nucleotide sequence(s) that encode the antibody, wherein said nucleotide sequence(s) are not naturally associated with the cell.
  • isolated protein “isolated polypeptide” or “isolated antibody” refers to a protein, polypeptide or antibody that by virtue of its origin or source of derivation (1 ) is not associated with naturally associated components that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, and/or (4) does not occur in nature.
  • a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components.
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • affinity refers to a measure of the attraction between an antigen and an antibody.
  • the intrinsic attractiveness of the antibody for the antigen is typically expressed as the binding affinity equilibrium constant (KD) of a particular antibody-antigen interaction.
  • KD binding affinity equilibrium constant
  • An antibody is said to specifically bind to an antigen when the KD for the binding is ⁇ 1 pM, e.g., ⁇ 100 nM or ⁇ 10 nM.
  • a KD binding affinity constant can be measured, e.g., by surface plasmon resonance (BIAcoreTM) using the IBIS MX96 SPR system from IBIS Technologies or the Carterra LSA SPR platform, or by Bio-Layer Interferometry, for example using the OctetTM system from ForteBio.
  • epitope refers to a portion (determinant) of an antigen that specifically binds to an antibody or a related molecule such as a bi-specific binding molecule.
  • Epitopic determinants generally consist of chemically active surface groupings of molecules such as amino acids or carbohydrate or sugar side chains and generally have specific three- dimensional structural characteristics, as well as specific charge characteristics.
  • An epitope may be “linear” or “conformational.” In a linear epitope, all of the points of interaction between a protein (e.g., an antigen) and an interacting molecule (such as an antibody) occur linearly along the primary amino acid sequence of the protein.
  • an antibody to a linear epitope may be generated, e.g., by immunizing an animal with a peptide having the amino acid residues of the linear epitope.
  • An antibody to a conformational epitope may be generated, e.g., by immunizing an animal with a mini-domain containing the relevant amino acid residues of the conformational epitope.
  • An antibody to a particular epitope can also be generated, e.g., by immunizing an animal with the target molecule of interest or a relevant portion thereof, then screening for binding to the epitope.
  • test antibody if the test antibody is not able to bind to the target at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound by the antibody of the present disclosure.
  • This experiment can be performed using, e.g., ELISA, RIA, BIACORETM, SPR, Bio-Layer Interferometry or flow cytometry.
  • competition method described above i.e., determining if the known antibody blocks the test antibody and vice versa.
  • Such cross-competition experiments may be performed, e.g., using an IBIS MX96 or Carterra LSA SPR instrument or the OctetTM system.
  • antibody portion refers to one or more portions or fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that certain fragments of a full-length antibody can perform the antigen-binding function of the antibody.
  • binding fragments encompassed within the term “antigen-binding portion” include (i) a Fab fragment: a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment: a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) an Fd fragment consisting of the VH and CH1 domains; (iv) an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment, which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) capable of specifically binding to an antigen.
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH1 domains
  • a F(ab')2 fragment a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region
  • the two domains of the Fv fragment, VL and VH are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH domains pair to form monovalent molecules (known as single chain Fv (scFv)).
  • antigenbinding molecules comprising a VH and/or a VL.
  • the molecule may also comprise one or more of a CH1 , hinge, CH2, or CH3 region.
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • Diabodies are bivalent, bi-specific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen-binding sites.
  • Antibody portions such as Fab and F(ab')2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesin molecules can be obtained using standard recombinant DNA techniques, e.g., as described herein.
  • the class (isotype) and subclass of antibodies may be determined by any method known in the art.
  • the class and subclass of an antibody may be determined using antibodies that are specific for a particular class and subclass of antibody. Such antibodies are available commercially.
  • the class and subclass can be determined by ELISA or Western blot as well as other techniques.
  • the class and subclass may be determined by sequencing all or a portion of the constant regions of the heavy and/or light chains of the antibodies, comparing their amino acid sequences to the known amino acid sequences of various classes and subclasses of immunoglobulins, and determining the class and subclass of the antibodies.
  • a combination therapy or composition described herein comprises an anti-EGFR antibody or an antigen-binding portion thereof.
  • the anti-EGFR antibody is futuximab, modotuximab, or a variant of any of these, where the variant may contain, e.g., certain minimum amino acid changes relative to said antibody (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid changes, which may be, e.g., in the framework regions) without losing the antigen-binding specificity of the antibody.
  • the anti-EGFR antibody competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, futuximab, or modotuximab.
  • the anti-EGFR antibody comprises H-CDR1 -3 and L-CDR1 -3 of futuximab, or modotuximab.
  • the anti-EGFR antibody comprises a VH and a VL at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the VH and VL of futuximab, or modotuximab.
  • the anti-EGFR antibody comprises the VH and VL of futuximab, or modotuximab.
  • the anti-EGFR antibody comprises an HC and an LC at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the HC and LC of futuximab, or modotuximab.
  • the anti-EGFR antibody comprises the HC and LC of futuximab, or modotuximab.
  • the anti-EGFR antibody comprises H-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 1 -3, or 11 -13.
  • the anti-EGFR antibody has a VH that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 7, or 17. In certain embodiments, the anti-EGFR antibody has a VH that comprises the amino acid sequence of SEQ ID NO: 7, or 17.
  • the anti-EGFR antibody has an HC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 9, or 19.
  • the anti-EGFR antibody has an HC that comprises the amino acid sequence of SEQ ID NO: 9, or 19.
  • the anti-EGFR antibody comprises L-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 4-6, or 14-16.
  • the anti-EGFR antibody has a VL that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 8, or 18. In certain embodiments, the anti-EGFR antibody has a VL that comprises the amino acid sequence of SEQ ID NO: 8, or 18.
  • the anti-EGFR antibody has an LC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 10, or 20. In certain embodiments, the anti-EGFR antibody has an LC that comprises the amino acid sequence of SEQ ID NO: 10, or 20.
  • the anti-EGFR antibody comprises any of the above heavy chain sequences and any of the above light chain sequences.
  • the anti-EGFR antibody comprises the H- CDR1 -3 and L-CDR1 -3 amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively.
  • the anti-EGFR antibody comprises a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
  • the anti-EGFR antibody comprises a VH and a VL that comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
  • the anti-EGFR antibody comprises an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
  • the anti-EGFR antibody comprises an HC and an LC that comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
  • an anti-EGFR combination of futuximab and modotuximab e.g., in a 1 : 1 ratio
  • an anti-EGFR antibody e.g., an anti-EGFR antibody or an anti-EGFR combination
  • an anti-EGFR component e.g., an anti-EGFR component
  • the anti-EGFR combination comprises first and second antibodies that compete or cross-compete for binding to human EGFR with, or bind to the same epitope of human EGFR as, futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise the H-CDR1 -3 and L-CDR1 -3 of futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise a VH and a VL at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the VH and VL of futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise the VH and VL of futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise an HC and an LC at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the HC and LC of futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise the HC and LC of futuximab and modotuximab, respectively.
  • the anti-EGFR combination comprises first and second antibodies with H-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 1-3 and SEQ ID NOs: 11 -13, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise a VH that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 7 and the amino acid sequence of SEQ ID NO: 17, respectively.
  • the anti- EGFR combination comprises a first antibody that has a VH that comprises the amino acid sequence of SEQ ID NO: 7, and a second antibody that has a VH that comprises the amino acid sequence of SEQ ID NO: 17.
  • the anti-EGFR combination comprises first and second antibodies that comprise an HC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 9 and the amino acid sequence of SEQ ID NO: 19, respectively.
  • the anti- EGFR combination comprises a first antibody that has an HC that comprises the amino acid sequence of SEQ ID NO: 9, and a second antibody that has an HC that comprises the amino acid sequence of SEQ ID NO: 19.
  • the anti-EGFR combination comprises first and second antibodies with L-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 4-6 and SEQ ID NOs: 14-16, respectively.
  • the anti-EGFR combination comprises first and second antibodies that comprise a VL that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 8 and the amino acid sequence of SEQ ID NO: 18, respectively.
  • the anti- EGFR combination comprises a first antibody that has a VL that comprises the amino acid sequence of SEQ ID NO: 8, and a second antibody that has a VL that comprises the amino acid sequence of SEQ ID NO: 18.
  • the anti-EGFR combination comprises first and second antibodies that comprise an LC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 10 and the amino acid sequence of SEQ ID NO: 20, respectively.
  • the anti- EGFR combination comprises a first antibody that has an LC that comprises the amino acid sequence of SEQ ID NO: 10 and a second antibody that has an LC that comprises the amino acid sequence of SEQ ID NO: 20.
  • the anti-EGFR combination comprises first and second antibodies with any of the above heavy chain sequences and any of the above light chain sequences for the first and second antibodies, respectively.
  • the anti-EGFR combination comprises: a) a first antibody comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 1 -6, respectively; and b) a second antibody comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 11-16, respectively.
  • the anti-EGFR combination comprises: a) a first antibody comprising a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 7 and 8, respectively; and b) a second antibody comprising a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 17 and 18, respectively.
  • the anti-EGFR combination comprises: a) a first antibody comprising a VH and a VL that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively; and b) a second antibody comprising a VH and a VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively.
  • the anti-EGFR combination comprises: a) a first antibody comprising an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 9 and 10, respectively; and b) a second antibody comprising an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
  • the anti-EGFR combination comprises: a) a first antibody comprising an HC and an LC that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively; and b) a second antibody comprising an HC and an LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
  • an anti-EGFR antibody or an antigen-binding portion thereof or an anti-EGFR combination as described herein is an anti-EGFR antibody or antigen-binding portion or combination described in PCT Patent Publication WO 2008/104183, which is incorporated by reference in its entirety herein.
  • nucleic acid molecule encoding VL or VH is isolated using methods well known in the art such that it does not include nucleic acid sequences encoding CL or CH, respectively.
  • the nucleic acid molecules encoding VL or VH then are operatively linked to a nucleic acid sequence encoding a CL or CH, respectively, from a different class of immunoglobulin molecule. This may be achieved using a vector or nucleic acid molecule that comprises a CL or CH sequence, as described above. For example, an antibody that was originally IgM may be class switched to IgG.
  • class switching may be used to convert one IgG subclass to another, e.g., from IgGi to lgG2.
  • a K light chain constant region can be changed, e.g., to a A light chain constant region, or vice-versa.
  • An exemplary method for producing an antibody described herein with a desired Ig isotype comprises the steps of isolating a nucleic acid molecule encoding the heavy chain of an antibody and a nucleic acid molecule encoding the light chain of an antibody, obtaining the variable domain of the heavy chain, ligating a coding sequence for the variable domain of the heavy chain with a coding sequence for the constant region of a heavy chain of the desired isotype, expressing the light chain and the heavy chain encoded by the ligated sequence in a cell, and collecting the antibody with the desired isotype.
  • An antibody described herein can be an IgG, an IgM, an IgE, an IgA, or an IgD molecule, but is typically of the IgG isotype, e.g., of IgG subclass IgGi, lgG 2a or lgG 2 b, IgGs or lgG4. In some embodiments, the antibody is of the isotype subclass IgGi .
  • the antibody may comprise at least one mutation in the Fc region.
  • a number of different Fc mutations are known, where these mutations alter the antibody’s effector function.
  • the antibody comprises at least one mutation in the Fc region that reduces effector function, e.g., mutations at one or more of positions 228, 233, 234 and 235, where amino acid positions are numbered according to Eu numbering.
  • one or both of the amino acid residues at positions 234 and 235 may be mutated, for example from Leu to Ala (L234A/L235A). These mutations reduce effector function of the Fc region of IgGi antibodies.
  • the amino acid positions are numbered according to the Eu numbering scheme.
  • the antibody may comprise the mutation S228P, where the amino acid position is numbered according to the Eu numbering scheme. This mutation is known to reduce undesired Fab arm exchange.
  • an antibody or antigen-binding portion thereof described herein may be part of a larger immunoadhesin molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • immunoadhesin molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov et al., Human Antibodies and Hybridomas (1995) 6:93-101 ) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov et al., Mol. Immunol.
  • CDRs from an antibody are incorporated into a molecule either covalently or noncovalently to make it an immunoadhesin that specifically binds to an antigen of interest.
  • the CDR(s) may be incorporated as part of a larger polypeptide chain, may be covalently linked to another polypeptide chain, or may be incorporated noncovalently.
  • a fusion antibody or immunoadhesin may be made that comprises all or a portion of an antibody described herein linked to another polypeptide.
  • only the variable domains of the antibody are linked to the polypeptide.
  • the VH domain of an antibody is linked to a first polypeptide, while the VL domain of an antibody is linked to a second polypeptide that associates with the first polypeptide in a manner such that the VH and VL domains can interact with one another to form an antigen-binding site.
  • the VH domain is separated from the VL domain by a linker such that the VH and VL domains can interact with one another (e.g., single-chain antibodies).
  • VH-linker-VL antibody is then linked to the polypeptide of interest.
  • fusion antibodies can be created in which two (or more) single-chain antibodies are linked to one another. This is useful if one wants to create a divalent or polyvalent antibody on a single polypeptide chain, or if one wants to create a bi-specific antibody.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3 (SEQ ID NO: 176), such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH domains joined by the flexible linker.
  • a flexible linker e.g., encoding the amino acid sequence (Gly4-Ser)3 (SEQ ID NO: 176)
  • the single chain antibody may be monovalent, if only a single VH and VL are used; bivalent, if two VH and VL are used; or polyvalent, if more than two VH and VL are used. Multispecific or polyvalent antibodies may be generated that bind specifically to the targets described herein, for instance.
  • modified antibodies may be prepared using antibody-encoding nucleic acid molecules.
  • “kappa bodies” III et al., Protein Eng. (1997) 10:949-57
  • “minibodies” Martin et al., EMBO J. (1994) 13:5303-9
  • “diabodies” Holliger et al., Proc. Natl. Acad. Sci. USA (1993) 90:6444-8
  • “Janusins” (Traunecker et al., EMBO J. (1991 ) 10:3655-9 and Traunecker et al., Int. J. Cancer (Suppl.) (1992) 7:51 - 2) may be prepared using standard molecular biological techniques following the teachings of the specification.
  • an antibody or antigen-binding portion described herein can be derivatized or linked to another molecule (e.g., another peptide or protein).
  • another molecule e.g., another peptide or protein.
  • the antibodies or portions thereof are derivatized such that target binding is not affected adversely by the derivatization or labeling. Accordingly, the antibodies and antibody portions of the present disclosure are intended to include both intact and modified forms of the antibodies described herein.
  • an antibody or antibody portion of the present disclosure can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bi-specific antibody or a diabody), a detection agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • another antibody e.g., a bi-specific antibody or a diabody
  • a detection agent e.g., a bi-specific antibody or a diabody
  • a pharmaceutical agent e.g., a drug that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies).
  • Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate).
  • Such linkers are available, e.g., from Pierce Chemical Company, Rockford, IL.
  • An antibody or antigen-binding portion can also be derivatized with a chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups may be useful to improve the biological characteristics of the antibody, e.g., to increase serum half-life.
  • PEG polyethylene glycol
  • an antibody or antigen-binding portion described herein may also be labeled.
  • the terms “label” or “labeled” refer to incorporation of another molecule in the antibody.
  • the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • the label or marker can be therapeutic, e.g., a drug conjugate or toxin.
  • Various methods of labeling polypeptides and glycoproteins are known in the art and may be used.
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 1111n, 1251, 1311), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, [3-galactosidase, luciferase, alkaline phosphatase), chemiluminescent markers, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), magnetic agents such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine
  • an antibody or antigen-binding portion described herein may be conjugated to a cytotoxic agent to form an immunoconjugate.
  • an antibody or antigen-binding portion according to the present disclosure may be conjugated to a radioisotope.
  • the antibodies described herein may be present in a neutral form (including zwitterionic forms) or as a positively or negatively-charged species.
  • the antibodies may be complexed with a counterion to form a pharmaceutically acceptable salt.
  • Trifl uridine/Ti pi racil composition Trifluridine (another name: a,a,a-trifluorothymidine) causes function inhibition of DNA by being incorporated into DNA of a tumor cell, and exhibits anti-tumor effects.
  • tipiracil hydrochloride (chemical name: 5-chloro-6-[(2-iminopyrrolidin-1 -yl)m ethyl]- pyri m idine-2 ,4( 1 /-/, 3/-/)- dione hydrochloride) has a thymidine phosphorylase inhibitory effect. It is known that tipiracil prevents in vivo degradation of trifluridine by thymidine phosphorylase, thus enhancing the anti-tumor effect of trifluridine (Investigational New Drugs, 26(5), 445-454, 2008).
  • an anti-tumor agent comprising trifluridine and tipiracil at a molar ratio of 1 :0.5
  • EMA/CHMP/130102/2016 a therapeutic agent for metastatic colorectal cancer which has been previously treated with available therapies including fluoropyrimidin, oxaliplatin and irinotecan based chemotherapies, anti-VEGF (Vascular Endothelial Growth Factor) agents and anti-EGFR (Epidermal Growth Factor receptor) agents.
  • Trifluridine/tipiracil drug and its use has been described for example in patent EP763529.
  • the present disclosure provides a combination therapy that comprises an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof in combination with a trifluridine/tipiracil component.
  • the combination therapy may take the form of, e.g., a method for treatment using said anti-EGFR antibody component or antigen-binding portions and trifluridine/tipiracil component or a pharmaceutical composition comprising said anti-EGFR antibody component or antigen-binding portions and trifluridine/tipiracil component.
  • the combination therapy or composition of the present disclosure comprises: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein (e.g., futuximab, modotuximab, or futuximab + modotuximab); b) a trifluridine/tipiracil component.
  • an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein (e.g., futuximab, modotuximab, or futuximab + modotuximab); b) a trifluridine/tipiracil component.
  • Sequences for the above-referenced antibodies may be, e.g., those found in Table 3.
  • the SEQ ID NOs for these sequences are assigned as shown in Table 1 below:
  • nucleic acid molecules and sequences antibodies or antigen-binding portions thereof described herein are also described.
  • different nucleic acid molecules encode the heavy chain and light chain amino acid sequences of the antibodies or antigen-binding portions.
  • the same nucleic acid molecule encodes the heavy chain and light chain amino acid sequences of the antibodies or antigen-binding portions.
  • a reference to a nucleotide sequence encompasses its complement unless otherwise specified. Thus, a reference to a nucleic acid having a particular sequence should be understood to encompass its complementary strand, with its complementary sequence.
  • polynucleotide as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes single- and double-stranded forms.
  • the present disclosure provides a nucleic acid molecule comprising a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, or a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of an antibody or antigen-binding portion thereof described herein.
  • the present disclosure also provides nucleotide sequences that are at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to a nucleotide sequence encoding an amino acid sequence selected from the group consisting of SEQ ID NOs: 7-10, 17-20.
  • the term “percent sequence identity” in the context of nucleic acid sequences refers to the residues in two sequences that are the same when aligned for maximum correspondence.
  • the length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 18 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36, 48 or more nucleotides.
  • FASTA Altschul et al.
  • FASTA which includes, e.g., the programs FASTA2 and FASTA3, provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (see, e.g., Pearson, Methods Enzymol. (1990) 183:63-98; Pearson, Methods Mol. Biol. (2000) 132:185-219; Pearson, Methods Enzymol. (1996) 266:227-58; and Pearson, J. Mol. Biol. (1998) 276:71 -84; incorporated herein by reference). Unless otherwise specified, default parameters for a particular program or algorithm are used.
  • percent sequence identity between nucleic acid sequences can be determined using FASTA with its default parameters (a word size of 6 and the NOPAM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1 , incorporated herein by reference.
  • nucleic acid molecules may be isolated.
  • Nucleic acid molecules referred to herein as “isolated” or “purified” are nucleic acids which (1 ) have been separated away from the nucleic acids of the genomic DNA or cellular RNA of their source of origin; and/or (2) do not occur in nature.
  • the present disclosure provides a vector suitable for expressing one or both of the chains of an antibody or antigen-binding portion thereof as described herein.
  • the term “vector”, as used herein, means a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • the vector is a plasmid, i.e., a circular double stranded piece of DNA into which additional DNA segments may be ligated.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • the present disclosure provides vectors comprising nucleic acid molecules that encode the heavy chain, the light chain, or both the heavy and light chains of an antibody as described herein or an antigen-binding portion thereof.
  • a vector of the present disclosure comprises a nucleic acid molecule described herein.
  • the present disclosure further provides vectors comprising nucleic acid molecules encoding fusion proteins, modified antibodies, antibody fragments, and probes thereof.
  • the vector may further comprise an expression control sequence.
  • expression control sequence means polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated.
  • Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • a nucleic acid molecule as described herein comprises a nucleotide sequence encoding a VH domain from an antibody or antigen-binding portion as described herein joined in-frame to a nucleotide sequence encoding a heavy chain constant region from any source.
  • a nucleic acid molecule as described herein can comprise a nucleotide sequence encoding a VL domain from an antibody or antigenbinding portion as described herein joined in-frame to a nucleotide sequence encoding a light chain constant region from any source.
  • nucleic acid molecules encoding the VH and/or VL may be “converted” to full-length antibody genes.
  • nucleic acid molecules encoding the VH or VL domains are converted to full-length antibody genes by insertion into an expression vector already encoding heavy chain constant (CH) or light chain constant (CL) regions, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector, and/or the VL segment is operatively linked to the CL segment within the vector.
  • CH heavy chain constant
  • CL light chain constant
  • nucleic acid molecules encoding the VH and/or VL domains are converted into full-length antibody genes by linking, e.g., ligating, a nucleic acid molecule encoding a VH and/or VL domain to a nucleic acid molecule encoding a CH and/or CL region using standard molecular biological techniques. Nucleic acid molecules encoding the full-length heavy and/or light chains may then be expressed from a cell into which they have been introduced and the antibody isolated.
  • the framework region(s) are mutated so that the resulting framework region(s) have the amino acid sequence of the corresponding germline gene.
  • a mutation may be made in a framework region or constant region, e.g., to increase the half-life of the antibody. See, e.g., PCT Publication WO 00/09560.
  • a mutation in a framework region or constant region also can be made to alter the immunogenicity of the antibody, and/or to provide a site for covalent or non-covalent binding to another molecule.
  • an antibody may have mutations in any one or more of the CDRs or framework regions of the variable domain or in the constant region.
  • One embodiment relates to a method for producing antibodies as described herein, comprising providing recombinant host cells capable of expressing the antibodies, culturing said host cells under conditions suitable for expression of the antibodies, and isolating the resulting antibodies.
  • Antibodies produced by such expression in such recombinant host cells are referred to herein as “recombinant antibodies.”
  • progeny cells of such host cells, and antibodies produced by same are also described.
  • recombinant host cell means a cell into which a recombinant expression vector has been introduced. By definition, a recombinant host cell does not occur in nature.
  • the present disclosure provides host cells that may comprise, e.g., a vector as described herein.
  • the present disclosure also provides host cells that comprise, e.g., a nucleotide sequence encoding the heavy chain or an antigen-binding portion thereof, a nucleotide sequence encoding the light chain or an antigen-binding portion thereof, or both, of an antibody or antigenbinding portion thereof described herein.
  • progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • Nucleic acid molecules encoding antibodies or antigen-binding portions thereof described herein and vectors comprising these nucleic acid molecules can be used for transfection of a suitable mammalian, plant, bacterial or yeast host cell. Transformation can be by any known method for introducing polynucleotides into a host cell. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors.
  • the present disclosure relates to a method for producing an antibody composition comprising one or two anti-EGFR antibodies or antigen-binding portions thereof, the method comprising:
  • first host cell is capable of expressing a first anti-EGFR antibody or an antigen-binding portion thereof as described herein and the second host cell is capable of expressing a second anti-EGFR antibody or antigen-binding portion thereof as described herein,
  • the present disclosure also provide host cells comprising: a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of a first anti-EGFR antibody or antigen-binding portion as described herein, and a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of a second anti-EGFR antibody as described herein;
  • Pharmaceutical Compositions comprising: a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of a first anti-EGFR antibody or antigen-binding portion as described herein, and a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof,
  • compositions comprising as active ingredients (e.g., as the sole active ingredients):
  • an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof; or I and
  • compositions comprising as an active ingredient (or as the sole active ingredient) a monotherapy or combination therapy of the present disclosure.
  • the pharmaceutical composition may additionally comprise a pharmaceutically acceptable excipient.
  • the pharmaceutical compositions are intended for amelioration, prevention, and/or treatment of cancer, e.g., a cancer described herein.
  • the cancer is in a tissue such as skin, lung, intestine, colon, ovary, brain, prostate, kidney, soft tissues, the hematopoietic system, head and neck, liver, bone, bladder, breast, stomach, uterus, cervix, and pancreas.
  • compositions of the present disclosure will comprise one or more antibodies, antigen-binding portions, a trifluridine/tipiracil composition, as described herein.
  • the composition comprises a trifluridine/tipiracil composition.
  • the composition comprises two antibodies described herein or antigen-binding portions thereof.
  • the composition comprises two antibodies as described herein or antigen-binding portions thereof and a trifluridine/tipiracil composition.
  • the composition comprises a monotherapy or combination therapy described herein.
  • the pharmaceutical composition may comprise a monotherapy or combination therapy of the present disclosure, and one or more additional agents selected from, e.g., an immunostimulatory agent, a vaccine, a chemotherapeutic agent, an anti-neoplastic agent, an anti- angiogenic agent, and a tyrosine kinase inhibitor.
  • the pharmaceutical composition is intended for amelioration, prevention, and/or treatment of a disorder, disease, or condition that improves, or slows down in its progression, by modulation of EGFR, phosphorylase inhibitory effect, inhibition of DNA by incorporation into DNA of a tumor cell or any combination thereof.
  • the pharmaceutical composition is intended for amelioration, prevention, and/or treatment of cancer.
  • the pharmaceutical composition is intended for activation of the immune system.
  • the therapies and compositions of the present disclosure are suitable to be administered as one or more formulations in association with one or more pharmaceutically acceptable excipient(s), e.g., as described below.
  • excipient is used herein to describe any ingredient other than the compound(s) of the present disclosure.
  • the choice of excipient(s) will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • pharmaceutically acceptable excipient includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • Some examples of pharmaceutically acceptable excipients are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • additional examples of pharmaceutically acceptable substances are wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • compositions of the present disclosure and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington’s Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995). Pharmaceutical compositions are preferably manufactured under GMP (good manufacturing practices) conditions.
  • a pharmaceutical composition of the present disclosure may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses.
  • a “unit dose” is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • Formulations of a pharmaceutical composition suitable for parenteral administration typically comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and the like. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline.
  • Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • injectable formulations may be prepared, packaged, or sold in unit dosage
  • the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • Parenteral formulations also include aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • Exemplary parenteral administration forms include solutions or suspensions in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, or in a liposomal preparation.
  • compositions according to the invention there may be mentioned more especially those that are suitable for administration by the oral, per- or trans-cutaneous, nasal, rectal, perlingual, ocular or respiratory route and more specifically tablets, dragees, sublingual tablets, gelatin capsules, glossettes, capsules, lozenges, aerosols, eye or nasal drops, suppositories, creams, ointments, dermal gels.
  • compositions according to the invention comprise one or more excipients or carriers chosen from diluents, lubricants, binders, disintegrators, stabilisers, preservatives, absorbents, colourings, sweeteners, flavourings.
  • lactose lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, glycerine;
  • binders aluminium and magnesium silicate, starch, gelatine, tragacanthin, methylcellulose, sodium carboxymethylcellulose and polyvinylpyrrolidone;
  • the therapies and compositions of the present disclosure are used to enhance or activate the immune system in a patient (e.g., a mammal such as a human) in need thereof.
  • the patient is immune-suppressed.
  • a physician can boost the anti-cancer activity of a patient’s own immune system by administering a therapy or composition as described herein.
  • a physician can boost anti-tumor activity in a patient by administering a therapy or composition of the present disclosure, alone or in combination with other therapeutic agents (sequentially or concurrently).
  • the therapies or compositions of the present disclosure are for use in the treatment of cancer.
  • the cancer may be in one or more tissues such as skin, lung, intestine, colon, ovary, brain, prostate, kidney, soft tissues, the hematopoietic system, head and neck, liver, bone, bladder, breast, stomach, uterus, cervix, and pancreas.
  • cancers treated by the therapies and compositions of the present disclosure may include, e.g., melanoma (e.g., advanced or metastatic melanoma), skin basal cell cancer, glioblastoma, glioma, gliosarcoma, astrocytoma, meningioma, neuroblastoma, adrenocortical cancer, head and neck squamous cell cancer, oral cancer, salivary gland cancer, nasopharyngeal cancer, breast cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC), small cell lung cancer, and squamous cell lung cancer), esophageal cancer, gastroesophageal junction cancer, gastric cancer, gastrointestinal cancer, primary peritoneal cancer, liver cancer, hepatocellular carcinoma, biliary tract cancer, colon cancer, rectal cancer, colorectal carcinoma, ovarian cancer, fallopian tube cancer, bladder cancer, upper urinary tract cancer,
  • the cancer may be, e.g., at an early, intermediate, late, locally advanced, advanced, or metastatic stage, may be relapsed, and may be refractory to and/or intolerant of other therapeutics (e.g., other therapeutics directed to one or more targets of the therapy or composition, checkpoint inhibitors, or standard of care for the cancer) or there may be no standard therapy available.
  • the cancer may not be amenable to surgical intervention due to either medical contraindications or non-resectability of the tumor.
  • conditions treated by the therapies and compositions of the present disclosure may include, e.g., colorectal cancer.
  • the colorectal cancer is metastatic, locally advanced, or unresectable.
  • the gastric cancer is (1 ) unresectable, (2) locally advanced or metastatic, (3) EGFR + , or (4) any combination (e.g., all) of (1 )-(3).
  • the patient with the colorectal cancer may have received treatment at least two prior regimens with first line standard therapy (e.g., cytotoxic chemotherapy, fluoropyrimidines, irinotecan and oxaliplatin, anti-VEGF inhibitor such as bevacizumab, aflibercept, ramucirumab, regorafenib, anti-EGFR such as cetuximab, panitumumab).
  • first line standard therapy e.g., cytotoxic chemotherapy, fluoropyrimidines, irinotecan and oxaliplatin, anti-VEGF inhibitor such as bevacizumab, aflibercept, ramucirumab, regorafenib, anti-EGFR such as cetuximab, panitumumab).
  • the colorectal cancer is (1 ) metastatic, (2) not amenable to surgical intervention due to either medical contraindications or non-resectability of the tumor, (3) with microsatellite instability status as low per institutional guidelines or guidelines from the College of American Pathologists, (4) any combination (e.g., all) of (1 )-(3).
  • the patient with the colorectal cancer may be (i) without RAS (KRAS and NRAS) mutations in any of the following codons: codons 12 and 13 in exon 2, codons 59 and 61 in exon 3, and codons 117 and 146 in exon 4; and/or (ii) without BRAF V600E mutation.
  • a therapy or composition of the present disclosure e.g., futuximab + modotuximab + trifluridine + tipiracil
  • the therapies or compositions of the present disclosure may be used to treat a patient population as described in Example 1.
  • a therapy or composition described herein may inhibit tumor growth and/or induce tumor growth regression in vivo. In some embodiments, a therapy or composition described herein may slow down or reverse metastasis in a cancer patient. In some embodiments, a therapy or composition described herein may prolong survival of a cancer patient. Any combination of the above properties is also contemplated.
  • the therapies or compositions of the present disclosure may be used in the treatment of an immune disorder.
  • the therapies or compositions of the present disclosure may be used to treat a patient who is, or is at risk of being, immunocompromised (e.g., due to chemotherapeutic or radiation therapy).
  • the therapies or compositions may be used to expand stem cells in a patient after stem cell transplantation.
  • Treatment refers to a method of alleviating or abrogating a biological disorder and/or at least one of its attendant symptoms.
  • to “alleviate” a disease, disorder or condition means reducing the severity and/or occurrence frequency of the symptoms of the disease, disorder, or condition.
  • references herein to “treatment” include references to curative, palliative and prophylactic treatment.
  • “Therapeutically effective amount” refers to the amount of the therapeutic agent being administered that will relieve to some extent one or more of the symptoms of the disorder being treated.
  • a therapeutically effective amount of an anti-cancer therapeutic may, for example, result in delayed tumor growth, tumor shrinkage, increased survival, elimination of cancer cells, slowed or decreased disease progression, reversal of metastasis, or other clinical endpoints desired by healthcare professionals.
  • a therapeutically effective amount of a therapy or composition of the present disclosure results in an improved objective response rate, improved clinical benefit rate, improved duration of response, increased progression-free survival, and increased overall survival, e.g., in comparison to untreated patients.
  • a therapy as described herein is administered in a single composition.
  • the therapy e.g., a combination therapy
  • a combination therapy comprising anti-EGFR antibodies component and trifluridine/tipiracil component may involve administration of a single composition, a composition comprising two antibodies and a composition comprising trifluridine/tipiracil component, or a separate composition for each antibody.
  • the compositions can be administered simultaneously, sequentially, separately, or any combination thereof.
  • the therapies or compositions of the present disclosure may be administered without additional therapeutic treatments, i.e., as a stand-alone therapy (monotherapy).
  • treatment with the therapy or combination may include at least one additional therapeutic treatment, e.g., another immunostimulatory agent, an anti-cancer agent (e.g., a chemotherapeutic agent, an anti-neoplastic agent, an anti-angiogenic agent, or a tyrosine kinase inhibitor), or a vaccine (e.g., a tumor vaccine).
  • an anti-cancer agent e.g., a chemotherapeutic agent, an anti-neoplastic agent, an anti-angiogenic agent, or a tyrosine kinase inhibitor
  • a vaccine e.g., a tumor vaccine
  • the therapy or composition may be coadministered or formulated with another medication/drug for the treatment of cancer.
  • the additional therapeutic treatment may comprise, e.g., an immunostimulatory agent, a vaccine, a chemotherapeutic agent, an anti- neoplastic agent, an anti-angiogenic agent, a tyrosine kinase inhibitor, and/or radiation therapy.
  • the additional therapeutic treatment may comprise a different anti-cancer antibody.
  • compositions as described herein and at least one other agent may be used as a combination treatment for simultaneous, separate or successive administration in cancer therapy.
  • agent e.g., a chemotherapeutic, anti- neoplastic, or anti-angiogenic agent
  • the other agent may by any agent suitable for treatment of the particular cancer in question, for example, an agent selected from the group consisting of alkylating agents, e.g., platinum derivatives such as cisplatin, carboplatin and/or oxaliplatin; plant alkoids, e.g., paclitaxel, docetaxel and/or irinotecan; antitumor antibiotics, e.g., doxorubicin (adriamycin), daunorubicin, epirubicin, idarubicin mitoxantrone, dactinomycin, bleomycin, actinomycin, luteomycin, and/or mitomycin; topoisomerase inhibitors such as topotecan; antimetabolites, e.g., fluorouracil and/or other fluoropyrimidines; FOLFOX; osimertinib; cyclophosphamide; anthracycline; dacarba
  • a therapy or composition of the present disclosure may also be used in combination with other anti-cancer therapies such as vaccines, cytokines, enzyme inhibitors, immunostimulatory compounds, and T cell therapies.
  • a vaccine it may be, e.g., a protein, peptide, or DNA vaccine containing one or more antigens which are relevant for the cancer being treated, or a vaccine comprising dendritic cells along with an antigen.
  • Suitable cytokines include, for example, IL-2, IFN-gamma and GM-CSF.
  • adoptive T cell therapy which refers to various immunotherapy techniques that involve expanding or engineering patients’ own T cells to recognize and attack their tumors.
  • a therapy or composition of the present disclosure may be used in adjunctive therapy in connection with tyrosine kinase inhibitors.
  • tyrosine kinase inhibitors synthetic, mainly quinazoline-derived, low molecular weight molecules that interact with the intracellular tyrosine kinase domain of receptors and inhibit ligand-induced receptor phosphorylation, e.g., by competing for the intracellular Mg-ATP binding site.
  • the therapy or composition may be used in combination with a medication/drug that mediates immune system activation, including, but not limited to, an agent that modulates the expression or activity of A2AR, A1AR, A2BR, A3AR, ADA, ALP, AXL, BTLA, B7-H3, B7- H4, CTLA-4, CD116, CD123, CD27, CD28, CD39, CD40, CD47, CD55, CD73, CD122, CD137, CD160, CGEN-15049, CHK1 , CHK2, CTLA-3, CEACAM (e.g., CEACAM-1 and/or CEACAM-5), EGFR, FLT3, HER2, NKG2AL, GAL9, GITR, HVEM, LAG-3, LILRB1 , LY108, LAIR1 , MET, NKG2A, ICOS, IDO, IL2R, IL4R, KIR, LAIR1 , PAP, PD-1/PD-L1/
  • the agent is a small molecule inhibitor.
  • the agent is an antibody or an antigen-binding fragment thereof that binds to one of the above molecules. It is also contemplated that a therapy or composition of the present disclosure may be used in combination with a cytokine (e.g., IL-1 , IL-2, IL-12, IL-15 or IL-21 ), a VEGF inhibitor.
  • the terms “co-administration,” “co-administered” and “in combination with,” referring to the therapies and compositions of the present disclosure with one or more other therapeutic agents, is intended to mean, and does refer to and include the following: a) simultaneous administration of such therapy/com position of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components at substantially the same time to said patient, b) substantially simultaneous administration of such therapy/composition of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at substantially the same time by said patient, whereupon said components are released at substantially the same time to said patient, c) sequential administration of such therapy/composition of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at consecutive times by said patient with a significant time interval between each administration, whereup
  • therapies and compositions of the present disclosure may be used in a method of treatment as described herein, may be for use in a treatment as described herein, and/or may be for use in the manufacture of a medicament for a treatment as described herein.
  • the therapies and compositions of the present disclosure may be administered in an effective amount for treatment of the condition in question, i.e. , at dosages and for periods of time necessary to achieve a desired result.
  • a therapeutically effective amount may vary according to factors such as the particular condition being treated, the age, sex and weight of the patient, and whether the antibodies are being administered as a stand-alone treatment or in combination with one or more additional anti-cancer treatments.
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the patients/subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the dose and dosing regimen are adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present disclosure.
  • dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the embodied composition. Further, the dosage regimen with the compositions of the present disclosure may be based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the seventy of the condition, the route of administration, and the particular antibody employed. Thus, the dosage regimen can vary widely, but can be determined routinely using standard methods.
  • doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values.
  • the present disclosure encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining of appropriate dosages and regimens is well- known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
  • An effective amount for tumor therapy may be measured by its ability to stabilize disease progression and/or ameliorate symptoms in a patient, and preferably to reverse disease progression, e.g., by reducing tumor size.
  • the ability of a therapy or composition of the present disclosure to inhibit cancer may be evaluated by in vitro assays, e.g., as described in the examples, as well as in suitable animal models that are predictive of the efficacy in human tumors.
  • Suitable dosage regimens will be selected in order to provide an optimum therapeutic response in each particular situation, for example, administered as a single bolus or as a continuous infusion, and with possible adjustment of the dosage as indicated by the exigencies of each case.
  • parenteral administration includes any route of administration characterized by physical breaching of a tissue of a subject and administration through the breach in the tissue, thus generally resulting in the direct administration into the blood stream, into muscle, or into an internal organ. Parenteral administration thus includes, but is not limited to, administration by injection, by application through a surgical incision, by application through a tissue-penetrating non-surgical wound, and the like.
  • parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal, intracisternal, intravenous, intraarterial, intrathecal, intraurethral, intracranial, intratumoral, and intrasynovial injection or infusions.
  • Particular embodiments include the intravenous and the subcutaneous routes.
  • the administration is IV injection, e.g., IV infusion.
  • the therapies and compositions of the present disclosure may be administered according to an exemplary dosing regimen described in Example 1.
  • the anti-EGFR antibodies component or antigen-binding portion may be administered at a dose of 1 , 3, 6, 9, 12, 15, or 18 mg/kg (e.g., as a monotherapy, or as part of a combination therapy, as described herein). ).
  • the anti-EGFR antibodies component or antigen-binding portion may be administered at one of said doses as a loading dose and a different one of said doses as a maintenance dose, for example a loading dose of 9 mg/kg followed by a maintenance dose of 6 mg/kg.
  • the anti-EGFR antibodies component or antigen-binding portion thereof is administered every 1 , 2, 3, or 4 weeks.
  • the anti-EGFR antibodies component or antigen-binding portion is administered first day every 1 , 2, 3, or 4, weeks. Further, 4 weeks constitutes a cycle of 28 days, the anti-EGFR antibodies component or antigen-binding portion is administered in cycle 1 , 2, 3 etc.
  • the trifluridine/tipiracil component may be administered at a dose of 35mg/m 2 twice daily (bid) (e.g., as part of a combination therapy as described herein).
  • the trifluridine/tipiracil component is administered during a cycle of 28 days on days 1 -5 of week 1 and days 1 -5 of week 2.
  • GGT Gamma-Glutamyl Transferase (Gamma-Glutamyl
  • IMP Investigational Medicinal Product a pharmaceutical form of an active ingredient or placebo being tested or used as a reference in a clinical trial (i.e. futuximab/modotuximab and trifluridine/tipiracil in this study)
  • TtPS2 Time to ECOG Performance Status > 2
  • TTR Time To Response
  • Example 1 Phase 3 clinical protocol for futuximab/modotuximab in combination therapy
  • This example describes a clinical trial protocol for an international, randomised, open label, multi-centre, two-arm Phase 3 study to evaluate futuximab/modotuximab in combination with trifluridine/tipiracil versus trifluridine/tipiracil monotherapy in approximately 500 patients with KRAS/NRAS and BRAF WT metastatic colorectal cancer (mCRC) who were previous treated by chemotherapy (including oxaliplatin, irinotecan and 5- fluorouracil, anti-VEGF agents) and with anti-EGFR mAb therapy for more than 4 months.
  • Maximum study duration for each participating patient each patient will participate in the study until confirmed disease progression, loss to followup, an adverse event leading to withdrawal, significant noncompliance with the study protocol, withdrawal of consent, end of study, or death from any cause.
  • a primary objective is to assess safety and tolerability of futuximab/modotuximab in combination with trifluridine/tipiracil.
  • the corresponding primary endpoints are:
  • DLTs dose-limiting toxicities
  • TEAEs Treatment Emergent Adverse Events
  • TESAEs Treatment Emergent Serious Adverse Events
  • a first secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil using Response Evaluation Criteria in Solid Tumours (RECIST) in terms of: Objective Response Rate (ORR), Best Overall Response (BOR), Disease Control Rate (DCR), Progression Free Survival (PFS).
  • RECIST Solid Tumours
  • ORR Objective Response Rate
  • BOR Best Overall Response
  • DCR Disease Control Rate
  • PFS Progression Free Survival
  • DC - Disease Control
  • PR disappearance of all target lesions
  • PR > 30% decrease in the sum of the longest diameter of target lesions
  • SD Stable Disease
  • a second secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil in terms of Overall Survival (OS).
  • OS Overall Survival
  • a third secondary objective is to characterise the pharmacokinetic (PK) profile of futuximab/modotuximab, trifluridine and tipiracil in the combination of futuximab/modotuximab with trifluridine/tipiracil.
  • the corresponding third secondary endpoint is :
  • the fourth secondary objective is to evaluate the immunogenicity of futuximab/modotuximab (i.e. occurrence of anti-drug antibody [ADA]).
  • the corresponding fourth secondary endpoint is :
  • the primary objective is to compare OS of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in patients with tumours that are KRAS/NRAS and BRAF WT (Double negative [DN]).
  • the corresponding primary endpoint is :
  • - OS (in DN) is defined as the time from date of randomisation into the study to death from any cause
  • the key secondary objective is to compare OS of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in patientswith tumours that are KRAS/NRAS, BRAF WT and EGFR- extracellular domain WT (Triple negative [TN]).
  • the corresponding key secondary endpoint is :
  • - OS (in DN) is defined as the time from date of randomisation into the study to death from any cause.
  • the first secondary objective is to compare anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy per Blinded Independent Central Review (BICR) assessment using RECIST in terms of Progression Free Survival (PFS), Objective Response Rate (ORR), Disease Control Rate (DCR), Duration of response (DoR) and Time to Response (TTR).
  • PFS Progression Free Survival
  • ORR Objective Response Rate
  • DCR Disease Control Rate
  • DoR Duration of response
  • TTR Time to Response
  • the second secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in terms of Time to Next Treatment (TTNT) and Time to ECOG Performance Status > 2 (TtPS2).
  • TTNT Time to Next Treatment
  • TtPS2 Time to ECOG Performance Status > 2
  • - TTNT the time from the randomisation to initiation of the next systemic anti-cancer therapy
  • TtPS2 the time from the date of randomization to the date when ECOG PS score of > 2 is observed for the first time
  • the third secondary objective is to further evaluate and to compare the safety profile of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy.
  • the corresponding third secondary endpoints are :
  • the fourth secondary objective is to compare Quality of life (QoL) of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy.
  • QoL Quality of life
  • the fifth secondary objective is to characterize the pharmacokinetic (PK) profile of futuximab/modotuximab, trifluridine and tipiracil in the combination futuximab/modotuximab with trifluridine/tipiracil.
  • PK pharmacokinetic
  • the sixth secondary objective is to further evaluate the immunogenicity of futuximab/modotuximab (/.e. occurrence of antidrug antibody [ADA]).
  • the corresponding first sixth endpoint is :
  • the study ( Figure 1 ) will comprise two parts a Safety Lead-In part (Figure 2) in approximately 25 patientsand a randomised Phase 3 part (Figure 3) in approximately 500 patients.
  • the Safety Lead-In part is an open-label, single-arm and non-random ised study phase in approximately 25 patients. During this part the safety and tolerability of futuximab/modotuximab in combination with trifluridine/tipiracil will be evaluated prior to the start of the Phase 3 part. Efficacy parameters, futuximab/modotuximab and trifluridine/tipiracil PK and futuximab/modotuximab immunogenicity will also be evaluated as secondary endpoints.
  • the therapeutic regimen for the Safety Lead-In part is as follows:
  • the Phase 3 part is of open-label, multi-centre, parallel-group, 2-arm, randomised design.
  • KRAS/NRAS and BRAF WT mCRC will be randomised in a 1 :1 ratio to either futuximab/modotuximab in combination with trifluridine/tipiracil (Arm A) or trifluridine/tipiracil (Arm B), and stratified according to performance status (PS) (0 versus 1 ), previous regimens of treatment (2 versus > 3) and by the presence or absence of EGFR ECD mutations.
  • Futuximab/modotuximab and trifluridine/tipiracil are the IMPs. Table 2 provides a description of the IMPs. Table 2
  • the medical and therapeutic criteria include :
  • fluoropyrimidines irinotecan and oxaliplatin
  • at least one anti-VEGF pathway inhibitor bevacizumab and/or aflibercept and/or ramucirumab and/or regorafenib
  • at least one anti-EGFR mAb cetuximab or panitumumab
  • the inclusion criteria include:
  • WOCBP Women of childbearing potential
  • tumour assessment will be done within 28 days prior to the first administration of first IMP.
  • tumour assessments will be done every 8 weeks from C1 D1 ( ⁇ 7 calendar days) until radiologic progression is documented.
  • tumour assessments will be done only if not performed within previous 8 weeks. Every effort should be made to perform the EOT tumour assessments prior to the start of new anti-cancer therapy.
  • tumour assessments will be done for patients who were withdrawn for reasons other than radiologic disease progression or consent withdrawal, every 8 weeks during the follow up period until the participant experienced radiologic progression, regardless of the initiation of a new anticancer therapy.
  • Tumour assessments will be performed as per RECIST version 1.1 at baseline and then every 8 weeks from C1 D1 ( ⁇ 7 calendar days) until radiologic progression, death or end of study, whichever occurs first.
  • the definition of responses for Target and Non-target lesions is presented below:
  • Compete Response the disappearance of all target lesions. Any pathological lymph nodes must have reduction in short axis to ⁇ 10 mm.
  • Partial Response at least a 30% decrease in the sum of diameters of the target lesions, taking as a reference the baseline sum diameters.
  • PD Progressive Disease: at least a 20% increase in the sum of diameters of the target lesions, taking as a reference the smallest sum on study, including the baseline sum. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. Definitive new lesion presence also indicates progression.
  • Stable Disease neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as a reference the smallest sum diameter while on study.
  • Compete Response The disappearance of all non-target lesions and normalisation of tumour marker level. All lymph nodes must be non- pathological morphologically (/.e., ⁇ 10 mm in short axis in size).
  • Non-CR/Non-PD A persistence of > 1 non-target lesion(s) and/or maintenance of tumour marker level above the normal limits (not reaching the extent of ‘unequivocal progression).
  • the investigator should consider all lesions (target and non-target) in assessing the tumor burden at repeat imaging prior to decide whether to continue treatment.
  • the participant has measurable disease and non- measurable disease, to achieve “unequivocal progression” on the basis of the non-target disease, there must be an overall level of substantial worsening in non-target disease such that, even in presence of SD or PR in target disease, the overall tumour burden has increased sufficiently to merit discontinuation of therapy.
  • a modest ‘increase’ in the size of one or more non-target lesions is usually not sufficient to quality for unequivocal progression status.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A combination comprising (a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof and (b) a trifluridine/tipiracil component.

Description

CANCER THERAPY TARGETING EGFR
BACKGROUND OF THE INVENTION
Cancer ranks as a leading cause of death and an important barrier to increasing life expectancy in every country of the world. According to estimates from the World Health Organization (WHO) in 2019, cancer is the first or second leading cause of death before the age of 70 years in 112 of 183 countries and ranks third or fourth in a further 23 countries.
Colorectal cancer (CRC) is the third most common cancer type in the world in terms of incidence and the second one in terms of mortality, with an estimated 1 ,800,000 new cases and 881 ,000 deaths in 2018.
Treatment for metastatic colorectal cancer (mCRC) is heavily dependent on gene expression, with many drugs being prescribed only for specific tumor profiles. Anti-PD-1 agents such as nivolumab and pembrolizumab have seen initial success in microsatellite instability high/defective mismatch repair (MSI-H/dMMR) mCRCs, although these comprise only 1 % of patients. Tumors that are RAS/BRAF wild-type (WT) and EGFR-positive are among the most common gene expression profiles, accounting for approximately 40% of CRCs.
Epidermal Growth Factor Receptor (EGFR) plays an important role in cellular proliferation as well as apoptosis, angiogenesis and metastatic spread, processes that are crucial to tumour progression (Salomon et al, Crit. Rev. Oncology/Haematology, 19:183-232 (1995); Wu et al, J. Clin. Invest., 95:1897-1905 (1995); Kames et al, Gastroenterology, 114:930-939 (1998); Woodburn et al, Pharmacol. Therap. 82: 241 -250 (1999); Price et al, Eur. J. Cancer, 32A: 1977-1982 (1996)). Indeed, studies have shown that EGFR- mediated cell growth is increased in a variety of solid tumours including nonsmall cell lung cancer, prostate cancer, breast cancer, gastric cancer, and tumours of the head and neck (Salomon DS et al, Critical Reviews in Oncology/Haematology, 19:183-232 (1995)). Furthermore, excessive activation of EGFR on the cancer cell surface is now known to be associated with advanced disease, the development of a metastatic phenotype and a poor prognosis in cancer patients (Salomon DS et al., Critical Reviews in Oncology/Haematology 19:183-232 (1995)).
In view of the critical situation of patients with mCRC becoming insensitive or unresponsive to first and second line chemotherapy, there is a need for new and improved therapies that combine anti-EGFR antibody and trifluridine/tipiracil to treat cancer.
SUMMARY OF THE INVENTION
The present invention is based on therapies for enhancing immunity comprising one or two anti-EGFR antibodies, e.g., as described herein, with trifluridine/tipiracil composition. In some embodiments, the therapy is for treating cancer. Also provided are pharmaceutical compositions comprising the components of the therapies, and use of the therapies for enhancing immunity (e.g., treating cancer) in a patient. The therapies described herein may be used in a method for enhancing immunity (e.g., treating cancer) in a patient; may be used for the manufacture of a medicament for enhancing immunity (e.g., treating cancer) in a patient; or may be for use in enhancing immunity (e.g., treating cancer) in a patient. By “enhancing immunity”, it is understood at least the blockage of the interaction between the ligandbinding domain of EGFR with EGFR ligands and concomitantly, the resultant intracellular signaling pathway. Indeed, EGFR expression is frequently accompanied by the production of EGFR-ligands, TGF-alpha and EGF among others, by EGFR-expressing tumour cells which suggests that an autocrine loop participates in the progression of these cells (Baselga, etal.(1994) Pharmac. Therapeut. 64: 127-154; Modjtahedi, et al. (1994) Int. J. Oncology. 4: 277-296). Blocking the interaction between such EGFR ligands and EGFR therefore can inhibit tumor growth and survival (Baselga, et al. (1994) Pharmac. Therapeut. 64: 127-154). Compared to currently available treatments (e.g., for cancer), it is contemplated that the therapies described herein may provide a superior clinical response.
In some embodiments, the present disclosure provides a combination comprising : a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof that competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component.
In certain embodiments, the heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 of the anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprise the amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively.
In some embodiments, the heavy chain variable domain (VH) and light chain variable domain (VL) of the anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
In certain embodiments, the heavy chain (HC) and light chain (LC) of the anti-EGFR component comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
In some embodiments, the heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 of an anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 1 -6, and the H-CDR1-3 and L-CDR1 -3 of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 11 -16.
In particular embodiments, the heavy chain variable domain (VH) and light chain variable domain (VL) of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 7 and 8, the VH and VL of the anti-EGFR antibody comprise the amino acid sequences of SEQ ID NOs: 17 and 18.
In some embodiments, The combination of claim 6, wherein the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
In some embodiments, the method of enhancing immunity in a human patient in need thereof comprises administering to the patient : a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof that competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component.
In certain embodiments, the anti-EGFR component comprises an anti- EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 that comprise the amino acid sequences of (a) SEQ ID NOs: 1 -6, respectively; or (b) SEQ ID NOs: 11 -16, respectively. In some embodiments, the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of (a) SEQ ID NOs: 7 and 8, respectively; or (b) SEQ ID NOs: 17 and 18, respectively. In particular embodiments, the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of (a) SEQ ID NOs: 9 and 10, respectively; or (b) SEQ ID NOs: 19 and 20, respectively.
In some embodiments, the method comprises administering the anti- EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H- CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1 -3 that comprise the amino acid sequences of SEQ ID NOs: 1 -6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with H- CDR1 -3 and L-CDR1 -3 that comprise the amino acid sequences of SEQ ID NOs: 11 -16, respectively.
In certain embodiment, the anti-EGFR component comprises an anti- EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti- EGFR antibody or an antigen-binding portion thereof with a VH and VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively. In particular embodiments, the anti-EGFR component comprises an anti- EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively. The anti-EGFR component may be for example futuximab, modotuximab, or futuximab + modotuximab.
In some embodiments, the method comprises administering to the patient: a) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 1-6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 11 -16, respectively; and a trifluridine/tipiracil component ; b) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 17 and 18, respectively; and a trifluridine/tipiracil component ; or c) an anti-EGFR component comprising an anti-EGFR antibody comprising the HC and LC amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody comprising the HC and LC amino acid sequences of SEQ ID NOs: 19 and 20, respectively; and a trifluridine/tipiracil component.
In any of the above methods, the one or two anti-EGFR antibodies or antigen-binding portions thereof and the trifluridine/tipiracil component may be administered to the patient concurrently or sequentially.
In some embodiments, the patient has cancer, e.g., a hematological malignancy or a solid tumor. In certain embodiments, the cancer is colorectal cancer or gastric cancer. In a preferred embodiment, the patient has a metastatic colorectal cancer (mCRC).
In some embodiments, the anti-EGFR component is administered at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, (e.g. weekly). In certain embodiments, the trifluridine/tipiracil component is administered at a dose of 20 to 80 mg/m2/day, optionally the component trifluridine/tipiracil is divided in two portions per day for five consecutive days and then a 2-days rest period. In certain embodiments, the trifluridine/tipiracil component is administered at a dose of 20mg/m2, 25mg/m2, 30mg/m2, 35mg/m2. In some embodiments, the one or two anti-EGFR antibodies or antigen-binding portions thereof are formulated for intravenous infusion. In some embodiments, the trifluridine/tipiracil component is formulated for orally administration. The present disclosure provides a method of treating an advanced solid tumor malignancy in a patient, comprising administering to the patient an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and a trifluridine/tipiracil component, wherein the anti-EGFR component is administered by IV infusion at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, wherein the trifluridine/tipiracil component is administered orally at a dose of 35mg/m2.
The present disclosure provides a method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; for example, the cancer is locally advanced and/or unresectable, and for example the patient has failed on first-line standard of care therapy and/or second-line standard of care therapy.
In some embodiments, the anti-EGFR antibody component is administered at a loading dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , and a maintenance dose of 6mg/kg day 8, day 15, day 22 of Cycle 1 each cycle and days 1 , 8 , 15, 22 of all other 28-days cycles, and the trifluridine/tipiracil component is administered at a dose of 35mg/m2 on days 1 to day 5 and days 8 to day 12 of each 28 days cycle, wherein the anti- EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally.
The present disclosure provides a method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; wherein the patient is (i) without RAS mutations in any of the following codons: codons 12 and 13 in exon 2, codons 59 and 61 in exon 3, and codons 117 and 146 in exon 4; and/or (ii) without the BRAF V600E mutation.
In certain embodiments, the anti-EGFR antibody component is administered at a loading dose dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , day 8, day 15, day 22 of each cycle, and the trifluridine/tipiracil component is administered at a dose of 35mg/m2 on days 1 to day 5 and days 8 to day 12 of each cycle, wherein the anti-EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally.
In some embodiments, a method of the present disclosure further comprises administering to the patient radiation therapy, or at least one of a chemotherapeutic agent, an anti-neoplastic agent, and an anti-angiogenic agent.
Treatment according to a method of the present disclosure may result in tumor regression, delay of tumor progression, inhibition of cancer progression, inhibition of cancer metastasis, prevention of cancer recurrence or residual disease, and/or prolonged survival. In some embodiments, treatment according to a method of the present disclosure may result in an improved objective response rate, improved clinical benefit rate, improved duration of response, increased progression-free survival, increased overall survival, or any combination thereof, e.g., in comparison to an untreated patient.
The present disclosure also provides a pharmaceutical composition comprising an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as described herein, and further comprising a trifluridine/tipiracil component, and a pharmaceutically acceptable excipient. The present disclosure also provides a pharmaceutical composition comprising the components of the method as described herein.
In particular embodiments, the pharmaceutical composition may comprise the antibodies or antigen-binding portions of any of the methods described herein, and may be for use in treating a human patient in any of the methods described herein.
The disclosure provides an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein for use in enhancing immunity in a human patient in need thereof in combination with a trifluridine/tipiracil component. In some embodiment, an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for use in treating a human patient in a method as described herein.
The present disclosure also provides use of an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for the manufacture of a medicament for enhancing immunity in a human patient in need thereof.
The present disclosure also provides use of an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein in combination with a trifluridine/tipiracil component for the manufacture of a medicament for treating a human patient in a method as described herein.
Other features, objectives, and advantages of the invention are apparent in the detailed description that follows. It should be understood, however, that the detailed description, while indicating embodiments and aspects of the invention, is given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art from the detailed description. BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1: study design
FIG.2 : study plan for the Safety Lead-In Part
FIG.3 : study plan for the Phase 3 Part
DETAILED DESCRIPTION OF THE INVENTION
The present disclosure provides new combination therapies and compositions that target human EGFR by using antibodies and trifluridine/tipiracil composition. The therapies (i.e., combination therapies) and compositions can be used to treat cancer in a human patient. Unless otherwise stated, as used herein, “EGFR” refers to the human forms of this target. A human EGFR polypeptide sequence is available under UniProt Accession No. P00533 (SEQ ID NO: 21 ).
The term “antibody” (Ab) or “immunoglobulin” (Ig), as used herein, refers to a tetramer comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa) inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable domain (VH) and a heavy chain constant region (CH). Each light chain is composed of a light chain variable domain (VL) and a light chain constant region (CL). The VH and VL domains can be subdivided further into regions of hypervariability, termed “complementarity determining regions” (CDRs), interspersed with regions that are more conserved, termed “framework regions” (FRs). Each VH and VL is composed of three CDRs (H-CDR herein designates a CDR from the heavy chain; and L-CDR herein designates a CDR from the light chain) and four FRs, arranged from amino-terminus to carboxyl-term inus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4. The assignment of amino acid numbers, and of FR and CDR regions, in the heavy or light chain may be in accordance with IMGT® definitions (Lefranc et al., Dev Comp Immunol (2003) 27(1):55-77); Eu numbering; or the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and 1991 )); Chothia & Lesk, J. Mol. Biol. (1987) 196:901-17; Chothia et al., Nature (1989) 342:878-83; MacCallum et al., J. Mol. Biol. (1996) 262:732-45; or Honegger and Pluckthun, J. Mol. Biol. (2001 ) 309(3):657-70.
The term “recombinant antibody” refers to an antibody that is expressed from a cell or cell line comprising the nucleotide sequence(s) that encode the antibody, wherein said nucleotide sequence(s) are not naturally associated with the cell.
The term “isolated protein” “isolated polypeptide” or “isolated antibody” refers to a protein, polypeptide or antibody that by virtue of its origin or source of derivation (1 ) is not associated with naturally associated components that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, and/or (4) does not occur in nature. Thus, a polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be “isolated” from its naturally associated components. A protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
The term “affinity” refers to a measure of the attraction between an antigen and an antibody. The intrinsic attractiveness of the antibody for the antigen is typically expressed as the binding affinity equilibrium constant (KD) of a particular antibody-antigen interaction. An antibody is said to specifically bind to an antigen when the KD for the binding is < 1 pM, e.g., < 100 nM or < 10 nM. A KD binding affinity constant can be measured, e.g., by surface plasmon resonance (BIAcore™) using the IBIS MX96 SPR system from IBIS Technologies or the Carterra LSA SPR platform, or by Bio-Layer Interferometry, for example using the Octet™ system from ForteBio.
The term “epitope” as used herein refers to a portion (determinant) of an antigen that specifically binds to an antibody or a related molecule such as a bi-specific binding molecule. Epitopic determinants generally consist of chemically active surface groupings of molecules such as amino acids or carbohydrate or sugar side chains and generally have specific three- dimensional structural characteristics, as well as specific charge characteristics. An epitope may be “linear” or “conformational.” In a linear epitope, all of the points of interaction between a protein (e.g., an antigen) and an interacting molecule (such as an antibody) occur linearly along the primary amino acid sequence of the protein. In a conformational epitope, the points of interaction occur across amino acid residues on the protein that are separated from one another in the primary amino acid sequence. Once a desired epitope on an antigen is determined, it is possible to generate antibodies to that epitope using techniques well known in the art. For example, an antibody to a linear epitope may be generated, e.g., by immunizing an animal with a peptide having the amino acid residues of the linear epitope. An antibody to a conformational epitope may be generated, e.g., by immunizing an animal with a mini-domain containing the relevant amino acid residues of the conformational epitope. An antibody to a particular epitope can also be generated, e.g., by immunizing an animal with the target molecule of interest or a relevant portion thereof, then screening for binding to the epitope.
One can determine whether an antibody binds to the same epitope as or competes for binding with an antibody as described herein by using methods known in the art, including, without limitation, competition assays, epitope binning, and alanine scanning. In some embodiments, one allows an antibody of the present disclosure to bind to its target under saturating conditions, and then measures the ability of the test antibody to bind to the target. If the test antibody is able to bind to the target at the same time as the reference antibody, then the test antibody binds to a different epitope than the reference antibody. However, if the test antibody is not able to bind to the target at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound by the antibody of the present disclosure. This experiment can be performed using, e.g., ELISA, RIA, BIACORE™, SPR, Bio-Layer Interferometry or flow cytometry. To test whether an antibody crosscompetes with another antibody, one may use the competition method described above in two directions, i.e., determining if the known antibody blocks the test antibody and vice versa. Such cross-competition experiments may be performed, e.g., using an IBIS MX96 or Carterra LSA SPR instrument or the Octet™ system.
The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more portions or fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that certain fragments of a full-length antibody can perform the antigen-binding function of the antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” include (i) a Fab fragment: a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment: a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) an Fd fragment consisting of the VH and CH1 domains; (iv) an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment, which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) capable of specifically binding to an antigen. Furthermore, although the two domains of the Fv fragment, VL and VH, are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH domains pair to form monovalent molecules (known as single chain Fv (scFv)). Also within the present disclosure are antigenbinding molecules comprising a VH and/or a VL. In the case of a VH, the molecule may also comprise one or more of a CH1 , hinge, CH2, or CH3 region. Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies, are also encompassed. Diabodies are bivalent, bi-specific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen-binding sites.
Antibody portions, such as Fab and F(ab')2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesin molecules can be obtained using standard recombinant DNA techniques, e.g., as described herein.
The class (isotype) and subclass of antibodies may be determined by any method known in the art. In general, the class and subclass of an antibody may be determined using antibodies that are specific for a particular class and subclass of antibody. Such antibodies are available commercially. The class and subclass can be determined by ELISA or Western blot as well as other techniques. Alternatively, the class and subclass may be determined by sequencing all or a portion of the constant regions of the heavy and/or light chains of the antibodies, comparing their amino acid sequences to the known amino acid sequences of various classes and subclasses of immunoglobulins, and determining the class and subclass of the antibodies.
Unless otherwise indicated, all antibody amino acid residue numbers referred to in this disclosure are those under the IMGT® numbering scheme. Anti-EGFR Antibodies
In some embodiments, a combination therapy or composition described herein comprises an anti-EGFR antibody or an antigen-binding portion thereof. In certain embodiments, the anti-EGFR antibody is futuximab, modotuximab, or a variant of any of these, where the variant may contain, e.g., certain minimum amino acid changes relative to said antibody (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid changes, which may be, e.g., in the framework regions) without losing the antigen-binding specificity of the antibody. In some embodiments, the anti-EGFR antibody competes or crosscompetes for binding to human EGFR with, or binds to the same epitope of human EGFR as, futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises H-CDR1 -3 and L-CDR1 -3 of futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises a VH and a VL at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the VH and VL of futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises the VH and VL of futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises an HC and an LC at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the HC and LC of futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises the HC and LC of futuximab, or modotuximab.
In some embodiments, the anti-EGFR antibody comprises H-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 1 -3, or 11 -13.
In some embodiments, the anti-EGFR antibody has a VH that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 7, or 17. In certain embodiments, the anti-EGFR antibody has a VH that comprises the amino acid sequence of SEQ ID NO: 7, or 17.
In some embodiments, the anti-EGFR antibody has an HC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 9, or 19. In certain embodiments, the anti-EGFR antibody has an HC that comprises the amino acid sequence of SEQ ID NO: 9, or 19. In some embodiments, the anti-EGFR antibody comprises L-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 4-6, or 14-16.
In some embodiments, the anti-EGFR antibody has a VL that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 8, or 18. In certain embodiments, the anti-EGFR antibody has a VL that comprises the amino acid sequence of SEQ ID NO: 8, or 18.
In some embodiments, the anti-EGFR antibody has an LC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 10, or 20. In certain embodiments, the anti-EGFR antibody has an LC that comprises the amino acid sequence of SEQ ID NO: 10, or 20.
In some embodiments, the anti-EGFR antibody comprises any of the above heavy chain sequences and any of the above light chain sequences.
In some embodiments, the anti-EGFR antibody comprises the H- CDR1 -3 and L-CDR1 -3 amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively.
In some embodiments, the anti-EGFR antibody comprises a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
In some embodiments, the anti-EGFR antibody comprises a VH and a VL that comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively.
In some embodiments, the anti-EGFR antibody comprises an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
In some embodiments, the anti-EGFR antibody comprises an HC and an LC that comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively.
In some embodiments, an anti-EGFR combination of futuximab and modotuximab (e.g., in a 1 : 1 ratio), or a combination comprising a variant of one or both antibodies, may be used where the combination therapy or composition of the present disclosure calls for an “anti-EGFR antibody.” In some embodiments, an anti-EGFR antibody or an anti-EGFR combination is referred to as an “anti-EGFR component.”
In some embodiments, the anti-EGFR combination comprises first and second antibodies that compete or cross-compete for binding to human EGFR with, or bind to the same epitope of human EGFR as, futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise the H-CDR1 -3 and L-CDR1 -3 of futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise a VH and a VL at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the VH and VL of futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise the VH and VL of futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise an HC and an LC at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the HC and LC of futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise the HC and LC of futuximab and modotuximab, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies with H-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 1-3 and SEQ ID NOs: 11 -13, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise a VH that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 7 and the amino acid sequence of SEQ ID NO: 17, respectively. In certain embodiments, the anti- EGFR combination comprises a first antibody that has a VH that comprises the amino acid sequence of SEQ ID NO: 7, and a second antibody that has a VH that comprises the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise an HC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 9 and the amino acid sequence of SEQ ID NO: 19, respectively. In certain embodiments, the anti- EGFR combination comprises a first antibody that has an HC that comprises the amino acid sequence of SEQ ID NO: 9, and a second antibody that has an HC that comprises the amino acid sequence of SEQ ID NO: 19.
In some embodiments, the anti-EGFR combination comprises first and second antibodies with L-CDR1 -3 comprising the amino acid sequences, respectively, of SEQ ID NOs: 4-6 and SEQ ID NOs: 14-16, respectively.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise a VL that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 8 and the amino acid sequence of SEQ ID NO: 18, respectively. In certain embodiments, the anti- EGFR combination comprises a first antibody that has a VL that comprises the amino acid sequence of SEQ ID NO: 8, and a second antibody that has a VL that comprises the amino acid sequence of SEQ ID NO: 18.
In some embodiments, the anti-EGFR combination comprises first and second antibodies that comprise an LC that is at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequence of SEQ ID NO: 10 and the amino acid sequence of SEQ ID NO: 20, respectively. In certain embodiments, the anti- EGFR combination comprises a first antibody that has an LC that comprises the amino acid sequence of SEQ ID NO: 10 and a second antibody that has an LC that comprises the amino acid sequence of SEQ ID NO: 20.
In some embodiments, the anti-EGFR combination comprises first and second antibodies with any of the above heavy chain sequences and any of the above light chain sequences for the first and second antibodies, respectively.
In some embodiments, the anti-EGFR combination comprises: a) a first antibody comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 1 -6, respectively; and b) a second antibody comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 11-16, respectively.
In some embodiments, the anti-EGFR combination comprises: a) a first antibody comprising a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 7 and 8, respectively; and b) a second antibody comprising a VH and a VL that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 17 and 18, respectively. In some embodiments, the anti-EGFR combination comprises: a) a first antibody comprising a VH and a VL that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively; and b) a second antibody comprising a VH and a VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively.
In some embodiments, the anti-EGFR combination comprises: a) a first antibody comprising an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 9 and 10, respectively; and b) a second antibody comprising an HC and an LC that are at least 90% (e.g., at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) identical to the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
In some embodiments, the anti-EGFR combination comprises: a) a first antibody comprising an HC and an LC that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively; and b) a second antibody comprising an HC and an LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively.
In some embodiments, an anti-EGFR antibody or an antigen-binding portion thereof or an anti-EGFR combination as described herein is an anti-EGFR antibody or antigen-binding portion or combination described in PCT Patent Publication WO 2008/104183, which is incorporated by reference in its entirety herein.
The class of an antibody described herein may be changed or switched with another class or subclass. In some embodiments of the present disclosure, a nucleic acid molecule encoding VL or VH is isolated using methods well known in the art such that it does not include nucleic acid sequences encoding CL or CH, respectively. The nucleic acid molecules encoding VL or VH then are operatively linked to a nucleic acid sequence encoding a CL or CH, respectively, from a different class of immunoglobulin molecule. This may be achieved using a vector or nucleic acid molecule that comprises a CL or CH sequence, as described above. For example, an antibody that was originally IgM may be class switched to IgG. Further, the class switching may be used to convert one IgG subclass to another, e.g., from IgGi to lgG2. A K light chain constant region can be changed, e.g., to a A light chain constant region, or vice-versa. An exemplary method for producing an antibody described herein with a desired Ig isotype comprises the steps of isolating a nucleic acid molecule encoding the heavy chain of an antibody and a nucleic acid molecule encoding the light chain of an antibody, obtaining the variable domain of the heavy chain, ligating a coding sequence for the variable domain of the heavy chain with a coding sequence for the constant region of a heavy chain of the desired isotype, expressing the light chain and the heavy chain encoded by the ligated sequence in a cell, and collecting the antibody with the desired isotype.
An antibody described herein can be an IgG, an IgM, an IgE, an IgA, or an IgD molecule, but is typically of the IgG isotype, e.g., of IgG subclass IgGi, lgG2a or lgG2b, IgGs or lgG4. In some embodiments, the antibody is of the isotype subclass IgGi .
In some embodiments, the antibody may comprise at least one mutation in the Fc region. A number of different Fc mutations are known, where these mutations alter the antibody’s effector function. For example, in some embodiments, the antibody comprises at least one mutation in the Fc region that reduces effector function, e.g., mutations at one or more of positions 228, 233, 234 and 235, where amino acid positions are numbered according to Eu numbering.
In some embodiments, e.g., where the antibody is of the IgGi subclass, one or both of the amino acid residues at positions 234 and 235 may be mutated, for example from Leu to Ala (L234A/L235A). These mutations reduce effector function of the Fc region of IgGi antibodies. The amino acid positions are numbered according to the Eu numbering scheme.
In some embodiments, e.g., where the antibody is of the lgG4 subclass, it may comprise the mutation S228P, where the amino acid position is numbered according to the Eu numbering scheme. This mutation is known to reduce undesired Fab arm exchange.
In certain embodiments, an antibody or antigen-binding portion thereof described herein may be part of a larger immunoadhesin molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesin molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov et al., Human Antibodies and Hybridomas (1995) 6:93-101 ) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov et al., Mol. Immunol. (1994) 31 :1047-58). Other examples include where one or more CDRs from an antibody are incorporated into a molecule either covalently or noncovalently to make it an immunoadhesin that specifically binds to an antigen of interest. In such embodiments, the CDR(s) may be incorporated as part of a larger polypeptide chain, may be covalently linked to another polypeptide chain, or may be incorporated noncovalently.
In another aspect, a fusion antibody or immunoadhesin may be made that comprises all or a portion of an antibody described herein linked to another polypeptide. In certain embodiments, only the variable domains of the antibody are linked to the polypeptide. In certain embodiments, the VH domain of an antibody is linked to a first polypeptide, while the VL domain of an antibody is linked to a second polypeptide that associates with the first polypeptide in a manner such that the VH and VL domains can interact with one another to form an antigen-binding site. In some embodiments, the VH domain is separated from the VL domain by a linker such that the VH and VL domains can interact with one another (e.g., single-chain antibodies). The VH-linker-VL antibody is then linked to the polypeptide of interest. In addition, fusion antibodies can be created in which two (or more) single-chain antibodies are linked to one another. This is useful if one wants to create a divalent or polyvalent antibody on a single polypeptide chain, or if one wants to create a bi-specific antibody.
To create a single chain antibody (scFv), the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3 (SEQ ID NO: 176), such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH domains joined by the flexible linker. See, e.g., Bird et al., Science (1988) 242:423-6; Huston et al., Proc. Natl. Acad. Sci. USA (1988) 85:5879-83; and McCafferty et al., Nature (1990) 348:552-4. The single chain antibody may be monovalent, if only a single VH and VL are used; bivalent, if two VH and VL are used; or polyvalent, if more than two VH and VL are used. Multispecific or polyvalent antibodies may be generated that bind specifically to the targets described herein, for instance.
In other embodiments, other modified antibodies may be prepared using antibody-encoding nucleic acid molecules. For instance, “kappa bodies” (III et al., Protein Eng. (1997) 10:949-57), “minibodies” (Martin et al., EMBO J. (1994) 13:5303-9), “diabodies” (Holliger et al., Proc. Natl. Acad. Sci. USA (1993) 90:6444-8), or “Janusins” (Traunecker et al., EMBO J. (1991 ) 10:3655-9 and Traunecker et al., Int. J. Cancer (Suppl.) (1992) 7:51 - 2) may be prepared using standard molecular biological techniques following the teachings of the specification.
An antibody or antigen-binding portion described herein can be derivatized or linked to another molecule (e.g., another peptide or protein). In general, the antibodies or portions thereof are derivatized such that target binding is not affected adversely by the derivatization or labeling. Accordingly, the antibodies and antibody portions of the present disclosure are intended to include both intact and modified forms of the antibodies described herein. For example, an antibody or antibody portion of the present disclosure can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bi-specific antibody or a diabody), a detection agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available, e.g., from Pierce Chemical Company, Rockford, IL.
An antibody or antigen-binding portion can also be derivatized with a chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups may be useful to improve the biological characteristics of the antibody, e.g., to increase serum half-life.
An antibody or antigen-binding portion described herein may also be labeled. As used herein, the terms “label” or “labeled” refer to incorporation of another molecule in the antibody. In some embodiments, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). In some embodiments, the label or marker can be therapeutic, e.g., a drug conjugate or toxin. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used. Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 1111n, 1251, 1311), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, [3-galactosidase, luciferase, alkaline phosphatase), chemiluminescent markers, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags), magnetic agents such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1 -dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
In some embodiments, an antibody or antigen-binding portion described herein may be conjugated to a cytotoxic agent to form an immunoconjugate. In some embodiments, an antibody or antigen-binding portion according to the present disclosure may be conjugated to a radioisotope.
In certain embodiments, the antibodies described herein may be present in a neutral form (including zwitterionic forms) or as a positively or negatively-charged species. In some embodiments, the antibodies may be complexed with a counterion to form a pharmaceutically acceptable salt. Trifl uridine/Ti pi racil composition Trifluridine (another name: a,a,a-trifluorothymidine) causes function inhibition of DNA by being incorporated into DNA of a tumor cell, and exhibits anti-tumor effects. Meanwhile, tipiracil hydrochloride (chemical name: 5-chloro-6-[(2-iminopyrrolidin-1 -yl)m ethyl]- pyri m idine-2 ,4( 1 /-/, 3/-/)- dione hydrochloride) has a thymidine phosphorylase inhibitory effect. It is known that tipiracil prevents in vivo degradation of trifluridine by thymidine phosphorylase, thus enhancing the anti-tumor effect of trifluridine (Investigational New Drugs, 26(5), 445-454, 2008). At the present time, an anti-tumor agent comprising trifluridine and tipiracil at a molar ratio of 1 :0.5 (hereinafter also called ’’trifluridine-tipiracil drug” or ’’TAS-102”) has been developed as a therapeutic agent of solid cancers and is approved in Japan as a therapeutic agent for unresectable advanced or recurrent colorectal cancer, and in United States and Europe under tradename Lonsurf® (EMA/CHMP/130102/2016) as a therapeutic agent for metastatic colorectal cancer which has been previously treated with available therapies including fluoropyrimidin, oxaliplatin and irinotecan based chemotherapies, anti-VEGF (Vascular Endothelial Growth Factor) agents and anti-EGFR (Epidermal Growth Factor receptor) agents.
Trifluridine/tipiracil drug and its use has been described for example in patent EP763529.
Combination Therapies
The present disclosure provides a combination therapy that comprises an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof in combination with a trifluridine/tipiracil component. The combination therapy may take the form of, e.g., a method for treatment using said anti-EGFR antibody component or antigen-binding portions and trifluridine/tipiracil component or a pharmaceutical composition comprising said anti-EGFR antibody component or antigen-binding portions and trifluridine/tipiracil component.
In some embodiments, the combination therapy or composition of the present disclosure comprises: a) an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as described herein (e.g., futuximab, modotuximab, or futuximab + modotuximab); b) a trifluridine/tipiracil component.
Sequences for the above-referenced antibodies may be, e.g., those found in Table 3. The SEQ ID NOs for these sequences are assigned as shown in Table 1 below:
Table 1. Antibody Sequence Identifiers
Figure imgf000027_0001
Nucleic Acid Molecules and Vectors
Also described are nucleic acid molecules and sequences antibodies or antigen-binding portions thereof described herein. In some embodiments, different nucleic acid molecules encode the heavy chain and light chain amino acid sequences of the antibodies or antigen-binding portions. In other embodiments, the same nucleic acid molecule encodes the heavy chain and light chain amino acid sequences of the antibodies or antigen-binding portions.
A reference to a nucleotide sequence encompasses its complement unless otherwise specified. Thus, a reference to a nucleic acid having a particular sequence should be understood to encompass its complementary strand, with its complementary sequence. The term “polynucleotide” as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes single- and double-stranded forms.
In some embodiments, the present disclosure provides a nucleic acid molecule comprising a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, or a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of an antibody or antigen-binding portion thereof described herein.
The present disclosure also provides nucleotide sequences that are at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to a nucleotide sequence encoding an amino acid sequence selected from the group consisting of SEQ ID NOs: 7-10, 17-20. The term “percent sequence identity” in the context of nucleic acid sequences refers to the residues in two sequences that are the same when aligned for maximum correspondence. The length of sequence identity comparison may be over a stretch of at least about nine nucleotides, usually at least about 18 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36, 48 or more nucleotides. There are a number of different algorithms known in the art which can be used to measure nucleotide sequence identity. For instance, polynucleotide sequences can be compared using FASTA, Gap or Bestfit, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wisconsin. FASTA, which includes, e.g., the programs FASTA2 and FASTA3, provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (see, e.g., Pearson, Methods Enzymol. (1990) 183:63-98; Pearson, Methods Mol. Biol. (2000) 132:185-219; Pearson, Methods Enzymol. (1996) 266:227-58; and Pearson, J. Mol. Biol. (1998) 276:71 -84; incorporated herein by reference). Unless otherwise specified, default parameters for a particular program or algorithm are used. For instance, percent sequence identity between nucleic acid sequences can be determined using FASTA with its default parameters (a word size of 6 and the NOPAM factor for the scoring matrix) or using Gap with its default parameters as provided in GCG Version 6.1 , incorporated herein by reference.
In any of the above embodiments, the nucleic acid molecules may be isolated. Nucleic acid molecules referred to herein as “isolated” or “purified” are nucleic acids which (1 ) have been separated away from the nucleic acids of the genomic DNA or cellular RNA of their source of origin; and/or (2) do not occur in nature.
In a further aspect, the present disclosure provides a vector suitable for expressing one or both of the chains of an antibody or antigen-binding portion thereof as described herein. The term “vector”, as used herein, means a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. In some embodiments, the vector is a plasmid, i.e., a circular double stranded piece of DNA into which additional DNA segments may be ligated. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
The present disclosure provides vectors comprising nucleic acid molecules that encode the heavy chain, the light chain, or both the heavy and light chains of an antibody as described herein or an antigen-binding portion thereof. In certain embodiments, a vector of the present disclosure comprises a nucleic acid molecule described herein. The present disclosure further provides vectors comprising nucleic acid molecules encoding fusion proteins, modified antibodies, antibody fragments, and probes thereof. The vector may further comprise an expression control sequence.
The term “expression control sequence” as used herein means polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence; in eukaryotes, generally, such control sequences include promoters and transcription termination sequence. The term “control sequences” is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
In some embodiments, a nucleic acid molecule as described herein comprises a nucleotide sequence encoding a VH domain from an antibody or antigen-binding portion as described herein joined in-frame to a nucleotide sequence encoding a heavy chain constant region from any source. Similarly, a nucleic acid molecule as described herein can comprise a nucleotide sequence encoding a VL domain from an antibody or antigenbinding portion as described herein joined in-frame to a nucleotide sequence encoding a light chain constant region from any source.
In a further aspect of the present disclosure, nucleic acid molecules encoding the VH and/or VL may be “converted” to full-length antibody genes. In some embodiments, nucleic acid molecules encoding the VH or VL domains are converted to full-length antibody genes by insertion into an expression vector already encoding heavy chain constant (CH) or light chain constant (CL) regions, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector, and/or the VL segment is operatively linked to the CL segment within the vector. In another aspect, nucleic acid molecules encoding the VH and/or VL domains are converted into full-length antibody genes by linking, e.g., ligating, a nucleic acid molecule encoding a VH and/or VL domain to a nucleic acid molecule encoding a CH and/or CL region using standard molecular biological techniques. Nucleic acid molecules encoding the full-length heavy and/or light chains may then be expressed from a cell into which they have been introduced and the antibody isolated.
In some embodiments, the framework region(s) are mutated so that the resulting framework region(s) have the amino acid sequence of the corresponding germline gene. A mutation may be made in a framework region or constant region, e.g., to increase the half-life of the antibody. See, e.g., PCT Publication WO 00/09560. A mutation in a framework region or constant region also can be made to alter the immunogenicity of the antibody, and/or to provide a site for covalent or non-covalent binding to another molecule. According to the present disclosure, an antibody may have mutations in any one or more of the CDRs or framework regions of the variable domain or in the constant region. Host Cells and Methods of Antibody and Antibody Composition Production
Also described are methods for producing the combination therapies (e.g., compositions) of the present disclosure. One embodiment relates to a method for producing antibodies as described herein, comprising providing recombinant host cells capable of expressing the antibodies, culturing said host cells under conditions suitable for expression of the antibodies, and isolating the resulting antibodies. Antibodies produced by such expression in such recombinant host cells are referred to herein as “recombinant antibodies.” Also described are progeny cells of such host cells, and antibodies produced by same.
The term “recombinant host cell” (or simply “host cell”), as used herein, means a cell into which a recombinant expression vector has been introduced. By definition, a recombinant host cell does not occur in nature. The present disclosure provides host cells that may comprise, e.g., a vector as described herein. The present disclosure also provides host cells that comprise, e.g., a nucleotide sequence encoding the heavy chain or an antigen-binding portion thereof, a nucleotide sequence encoding the light chain or an antigen-binding portion thereof, or both, of an antibody or antigenbinding portion thereof described herein. It should be understood that “recombinant host cell” and “host cell” mean not only the particular subject cell but also the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
Nucleic acid molecules encoding antibodies or antigen-binding portions thereof described herein and vectors comprising these nucleic acid molecules can be used for transfection of a suitable mammalian, plant, bacterial or yeast host cell. Transformation can be by any known method for introducing polynucleotides into a host cell. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors.
It is likely that antibodies expressed by different cell lines or in transgenic animals will have different glycosylation patterns from each other. However, all antibodies encoded by the nucleic acid molecules provided herein, or comprising the amino acid sequences provided herein are part of the present disclosure, regardless of the glycosylation state of the antibodies, and more generally, regardless of the presence or absence of post- translational modification(s).
In some embodiments, the present disclosure relates to a method for producing an antibody composition comprising one or two anti-EGFR antibodies or antigen-binding portions thereof, the method comprising:
- providing first and second host cells, wherein the first host cell is capable of expressing a first anti-EGFR antibody or an antigen-binding portion thereof as described herein and the second host cell is capable of expressing a second anti-EGFR antibody or antigen-binding portion thereof as described herein,
- cultivating the first and second host cells under conditions suitable for expression of the anti-EGFR antibodies, and
- isolating the resulting antibodies.
The present disclosure also provide host cells comprising: a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of a first anti-EGFR antibody or antigen-binding portion as described herein, and a nucleotide sequence that encodes the heavy chain or an antigen-binding portion thereof, a nucleotide sequence that encodes the light chain or an antigen-binding portion thereof, or both, of a second anti-EGFR antibody as described herein; Pharmaceutical Compositions
Another aspect of the present disclosure is a pharmaceutical composition comprising as active ingredients (e.g., as the sole active ingredients):
- an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof; or I and
- a trifluridine/tipiracil component.
Another aspect of the present disclosure is a pharmaceutical composition comprising as an active ingredient (or as the sole active ingredient) a monotherapy or combination therapy of the present disclosure. The pharmaceutical composition may additionally comprise a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical compositions are intended for amelioration, prevention, and/or treatment of cancer, e.g., a cancer described herein. In certain embodiments, the cancer is in a tissue such as skin, lung, intestine, colon, ovary, brain, prostate, kidney, soft tissues, the hematopoietic system, head and neck, liver, bone, bladder, breast, stomach, uterus, cervix, and pancreas.
Pharmaceutical compositions of the present disclosure will comprise one or more antibodies, antigen-binding portions, a trifluridine/tipiracil composition, as described herein. In some embodiments, the composition comprises a trifluridine/tipiracil composition. In some embodiments, the composition comprises two antibodies described herein or antigen-binding portions thereof. In some embodiments, the composition comprises two antibodies as described herein or antigen-binding portions thereof and a trifluridine/tipiracil composition. In some embodiments, the composition comprises a monotherapy or combination therapy described herein.
In some embodiments, the pharmaceutical composition may comprise a monotherapy or combination therapy of the present disclosure, and one or more additional agents selected from, e.g., an immunostimulatory agent, a vaccine, a chemotherapeutic agent, an anti-neoplastic agent, an anti- angiogenic agent, and a tyrosine kinase inhibitor. In some embodiments, the pharmaceutical composition is intended for amelioration, prevention, and/or treatment of a disorder, disease, or condition that improves, or slows down in its progression, by modulation of EGFR, phosphorylase inhibitory effect, inhibition of DNA by incorporation into DNA of a tumor cell or any combination thereof. In some embodiments, the pharmaceutical composition is intended for amelioration, prevention, and/or treatment of cancer. In some embodiments, the pharmaceutical composition is intended for activation of the immune system.
Generally, the therapies and compositions of the present disclosure are suitable to be administered as one or more formulations in association with one or more pharmaceutically acceptable excipient(s), e.g., as described below.
The term “excipient” is used herein to describe any ingredient other than the compound(s) of the present disclosure. The choice of excipient(s) will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form. As used herein, “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Some examples of pharmaceutically acceptable excipients are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Additional examples of pharmaceutically acceptable substances are wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
Pharmaceutical compositions of the present disclosure and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington’s Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995). Pharmaceutical compositions are preferably manufactured under GMP (good manufacturing practices) conditions.
A pharmaceutical composition of the present disclosure may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses. As used herein, a “unit dose” is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
Formulations of a pharmaceutical composition suitable for parenteral administration typically comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and the like. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents. In some embodiments of a formulation for parenteral administration, the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition. Parenteral formulations also include aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water. Exemplary parenteral administration forms include solutions or suspensions in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired. Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, or in a liposomal preparation.
Among the pharmaceutical compositions according to the invention there may be mentioned more especially those that are suitable for administration by the oral, per- or trans-cutaneous, nasal, rectal, perlingual, ocular or respiratory route and more specifically tablets, dragees, sublingual tablets, gelatin capsules, glossettes, capsules, lozenges, aerosols, eye or nasal drops, suppositories, creams, ointments, dermal gels.
In addition to the active principles, the pharmaceutical compositions according to the invention comprise one or more excipients or carriers chosen from diluents, lubricants, binders, disintegrators, stabilisers, preservatives, absorbents, colourings, sweeteners, flavourings.
Examples which may be mentioned, without implying any limitation, include:
- for the diluents: lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, glycerine;
- for the lubricants: silica, talc, stearic acid and its magnesium and calcium salts, polyethylene glycol;
- for the binders: aluminium and magnesium silicate, starch, gelatine, tragacanthin, methylcellulose, sodium carboxymethylcellulose and polyvinylpyrrolidone;
- for the disintegrators: agar, alginic acid and its sodium salt, effervescent mixtures. Therapeutic uses of antibodies and compositions of the present disclosure
In some embodiments, the therapies and compositions of the present disclosure are used to enhance or activate the immune system in a patient (e.g., a mammal such as a human) in need thereof. In certain embodiments, the patient is immune-suppressed. In certain embodiments, a physician can boost the anti-cancer activity of a patient’s own immune system by administering a therapy or composition as described herein. For example, a physician can boost anti-tumor activity in a patient by administering a therapy or composition of the present disclosure, alone or in combination with other therapeutic agents (sequentially or concurrently).
In certain embodiments, the therapies or compositions of the present disclosure are for use in the treatment of cancer. The cancer may be in one or more tissues such as skin, lung, intestine, colon, ovary, brain, prostate, kidney, soft tissues, the hematopoietic system, head and neck, liver, bone, bladder, breast, stomach, uterus, cervix, and pancreas.
In some embodiments, cancers treated by the therapies and compositions of the present disclosure may include, e.g., melanoma (e.g., advanced or metastatic melanoma), skin basal cell cancer, glioblastoma, glioma, gliosarcoma, astrocytoma, meningioma, neuroblastoma, adrenocortical cancer, head and neck squamous cell cancer, oral cancer, salivary gland cancer, nasopharyngeal cancer, breast cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC), small cell lung cancer, and squamous cell lung cancer), esophageal cancer, gastroesophageal junction cancer, gastric cancer, gastrointestinal cancer, primary peritoneal cancer, liver cancer, hepatocellular carcinoma, biliary tract cancer, colon cancer, rectal cancer, colorectal carcinoma, ovarian cancer, fallopian tube cancer, bladder cancer, upper urinary tract cancer, urothelial cancer, renal cell carcinoma, kidney cancer, genitourinary cancer, cervical cancer, prostate cancer, fibrosarcoma, liposarcoma, rhabdomyosarcoma, osteosarcoma, histiocytoma, pancreatic cancer, endometrial cancer, cancer of the appendix, advanced Merkel cell cancer, multiple myeloma, sarcomas, choriocarcinoma, erythroleukemia, acute lymphoblastic leukemia, acute monocytic leukemia, acute promyelocytic leukemia, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, acute lymphoblastic leukemia, mast cell leukemia, small lymphocytic lymphoma, Burkitt’s lymphoma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, diffuse large B cell lymphoma, follicular lymphoma, monocytic lymphoma, HTLV- associated T cell leukemia/lymphoma, mesothelioma, and solid tumors. The cancer may be, e.g., at an early, intermediate, late, locally advanced, advanced, or metastatic stage, may be relapsed, and may be refractory to and/or intolerant of other therapeutics (e.g., other therapeutics directed to one or more targets of the therapy or composition, checkpoint inhibitors, or standard of care for the cancer) or there may be no standard therapy available. In some embodiments, the cancer may not be amenable to surgical intervention due to either medical contraindications or non-resectability of the tumor.
In some embodiments, conditions treated by the therapies and compositions of the present disclosure may include, e.g., colorectal cancer. In some embodiments, the colorectal cancer is metastatic, locally advanced, or unresectable.
In some embodiments, the gastric cancer is (1 ) unresectable, (2) locally advanced or metastatic, (3) EGFR+, or (4) any combination (e.g., all) of (1 )-(3). Additionally or alternatively, the patient with the colorectal cancer may have received treatment at least two prior regimens with first line standard therapy (e.g., cytotoxic chemotherapy, fluoropyrimidines, irinotecan and oxaliplatin, anti-VEGF inhibitor such as bevacizumab, aflibercept, ramucirumab, regorafenib, anti-EGFR such as cetuximab, panitumumab).
In some embodiments, the colorectal cancer is (1 ) metastatic, (2) not amenable to surgical intervention due to either medical contraindications or non-resectability of the tumor, (3) with microsatellite instability status as low per institutional guidelines or guidelines from the College of American Pathologists, (4) any combination (e.g., all) of (1 )-(3). Additionally or alternatively, the patient with the colorectal cancer may be (i) without RAS (KRAS and NRAS) mutations in any of the following codons: codons 12 and 13 in exon 2, codons 59 and 61 in exon 3, and codons 117 and 146 in exon 4; and/or (ii) without BRAF V600E mutation. In certain embodiments, a therapy or composition of the present disclosure (e.g., futuximab + modotuximab + trifluridine + tipiracil) may be used to treat metastatic colorectal cancer.
In some embodiments, the therapies or compositions of the present disclosure may be used to treat a patient population as described in Example 1.
In some embodiments, a therapy or composition described herein may inhibit tumor growth and/or induce tumor growth regression in vivo. In some embodiments, a therapy or composition described herein may slow down or reverse metastasis in a cancer patient. In some embodiments, a therapy or composition described herein may prolong survival of a cancer patient. Any combination of the above properties is also contemplated.
In some embodiments, the therapies or compositions of the present disclosure may be used in the treatment of an immune disorder.
In some embodiments, the therapies or compositions of the present disclosure may be used to treat a patient who is, or is at risk of being, immunocompromised (e.g., due to chemotherapeutic or radiation therapy). In some embodiments, the therapies or compositions may be used to expand stem cells in a patient after stem cell transplantation.
“Treat,” “treating,” and “treatment” refer to a method of alleviating or abrogating a biological disorder and/or at least one of its attendant symptoms. As used herein, to “alleviate” a disease, disorder or condition means reducing the severity and/or occurrence frequency of the symptoms of the disease, disorder, or condition. Further, references herein to “treatment” include references to curative, palliative and prophylactic treatment.
“Therapeutically effective amount” refers to the amount of the therapeutic agent being administered that will relieve to some extent one or more of the symptoms of the disorder being treated. A therapeutically effective amount of an anti-cancer therapeutic may, for example, result in delayed tumor growth, tumor shrinkage, increased survival, elimination of cancer cells, slowed or decreased disease progression, reversal of metastasis, or other clinical endpoints desired by healthcare professionals. In some embodiments, a therapeutically effective amount of a therapy or composition of the present disclosure results in an improved objective response rate, improved clinical benefit rate, improved duration of response, increased progression-free survival, and increased overall survival, e.g., in comparison to untreated patients.
In some embodiments, a therapy as described herein is administered in a single composition. In other embodiments, the therapy (e.g., a combination therapy) is administered in more than one composition. For example, a combination therapy comprising anti-EGFR antibodies component and trifluridine/tipiracil component may involve administration of a single composition, a composition comprising two antibodies and a composition comprising trifluridine/tipiracil component, or a separate composition for each antibody. In a case where there is more than one composition, the compositions can be administered simultaneously, sequentially, separately, or any combination thereof.
The therapies or compositions of the present disclosure may be administered without additional therapeutic treatments, i.e., as a stand-alone therapy (monotherapy). Alternatively, treatment with the therapy or combination may include at least one additional therapeutic treatment, e.g., another immunostimulatory agent, an anti-cancer agent (e.g., a chemotherapeutic agent, an anti-neoplastic agent, an anti-angiogenic agent, or a tyrosine kinase inhibitor), or a vaccine (e.g., a tumor vaccine).
In some embodiments, the therapy or composition may be coadministered or formulated with another medication/drug for the treatment of cancer. The additional therapeutic treatment may comprise, e.g., an immunostimulatory agent, a vaccine, a chemotherapeutic agent, an anti- neoplastic agent, an anti-angiogenic agent, a tyrosine kinase inhibitor, and/or radiation therapy. In some embodiments, the additional therapeutic treatment may comprise a different anti-cancer antibody.
Pharmaceutical articles comprising a therapy or composition as described herein and at least one other agent (e.g., a chemotherapeutic, anti- neoplastic, or anti-angiogenic agent) may be used as a combination treatment for simultaneous, separate or successive administration in cancer therapy. The other agent may by any agent suitable for treatment of the particular cancer in question, for example, an agent selected from the group consisting of alkylating agents, e.g., platinum derivatives such as cisplatin, carboplatin and/or oxaliplatin; plant alkoids, e.g., paclitaxel, docetaxel and/or irinotecan; antitumor antibiotics, e.g., doxorubicin (adriamycin), daunorubicin, epirubicin, idarubicin mitoxantrone, dactinomycin, bleomycin, actinomycin, luteomycin, and/or mitomycin; topoisomerase inhibitors such as topotecan; antimetabolites, e.g., fluorouracil and/or other fluoropyrimidines; FOLFOX; osimertinib; cyclophosphamide; anthracycline; dacarbazine; gemcitabine; or any combination thereof. In some embodiments, the therapy or composition described herein reestablishes responsiveness to the other agent.
A therapy or composition of the present disclosure may also be used in combination with other anti-cancer therapies such as vaccines, cytokines, enzyme inhibitors, immunostimulatory compounds, and T cell therapies. In the case of a vaccine, it may be, e.g., a protein, peptide, or DNA vaccine containing one or more antigens which are relevant for the cancer being treated, or a vaccine comprising dendritic cells along with an antigen. Suitable cytokines include, for example, IL-2, IFN-gamma and GM-CSF. Also contemplated is adoptive T cell therapy, which refers to various immunotherapy techniques that involve expanding or engineering patients’ own T cells to recognize and attack their tumors.
It is also contemplated that a therapy or composition of the present disclosure may be used in adjunctive therapy in connection with tyrosine kinase inhibitors. These are synthetic, mainly quinazoline-derived, low molecular weight molecules that interact with the intracellular tyrosine kinase domain of receptors and inhibit ligand-induced receptor phosphorylation, e.g., by competing for the intracellular Mg-ATP binding site.
In some embodiments, the therapy or composition may be used in combination with a medication/drug that mediates immune system activation, including, but not limited to, an agent that modulates the expression or activity of A2AR, A1AR, A2BR, A3AR, ADA, ALP, AXL, BTLA, B7-H3, B7- H4, CTLA-4, CD116, CD123, CD27, CD28, CD39, CD40, CD47, CD55, CD73, CD122, CD137, CD160, CGEN-15049, CHK1 , CHK2, CTLA-3, CEACAM (e.g., CEACAM-1 and/or CEACAM-5), EGFR, FLT3, HER2, NKG2AL, GAL9, GITR, HVEM, LAG-3, LILRB1 , LY108, LAIR1 , MET, NKG2A, ICOS, IDO, IL2R, IL4R, KIR, LAIR1 , PAP, PD-1/PD-L1/PD-L2, 0X40, STING, TIGIT, TIM-3, TGFR-beta, TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9 and TLR10, TNFR2, VEGFR, VEGF, VISTA, LILRB2, CMTM6 and/or 2B4. In certain embodiments, the agent is a small molecule inhibitor. In certain embodiments, the agent is an antibody or an antigen-binding fragment thereof that binds to one of the above molecules. It is also contemplated that a therapy or composition of the present disclosure may be used in combination with a cytokine (e.g., IL-1 , IL-2, IL-12, IL-15 or IL-21 ), a VEGF inhibitor.
As used herein, the terms “co-administration,” “co-administered” and “in combination with,” referring to the therapies and compositions of the present disclosure with one or more other therapeutic agents, is intended to mean, and does refer to and include the following: a) simultaneous administration of such therapy/com position of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components at substantially the same time to said patient, b) substantially simultaneous administration of such therapy/composition of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at substantially the same time by said patient, whereupon said components are released at substantially the same time to said patient, c) sequential administration of such therapy/composition of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at consecutive times by said patient with a significant time interval between each administration, whereupon said components are released at substantially different times to said patient; and d) sequential administration of such therapy/composition of the present disclosure and therapeutic agent(s) to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components in a controlled manner whereupon they are concurrently, consecutively, and/or overlappingly released at the same and/or different times to said patient, where each part may be administered by either the same or a different route.
It is understood that the therapies and compositions of the present disclosure may be used in a method of treatment as described herein, may be for use in a treatment as described herein, and/or may be for use in the manufacture of a medicament for a treatment as described herein.
Dose and Route of Administration
The therapies and compositions of the present disclosure may be administered in an effective amount for treatment of the condition in question, i.e. , at dosages and for periods of time necessary to achieve a desired result. A therapeutically effective amount may vary according to factors such as the particular condition being treated, the age, sex and weight of the patient, and whether the antibodies are being administered as a stand-alone treatment or in combination with one or more additional anti-cancer treatments.
Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suited as unitary dosages for the patients/subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the present disclosure are generally dictated by and directly dependent on (a) the unique characteristics of the therapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
Thus, the skilled artisan would appreciate, based upon the disclosure provided herein, that the dose and dosing regimen are adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present disclosure.
It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the embodied composition. Further, the dosage regimen with the compositions of the present disclosure may be based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the seventy of the condition, the route of administration, and the particular antibody employed. Thus, the dosage regimen can vary widely, but can be determined routinely using standard methods. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the present disclosure encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining of appropriate dosages and regimens is well- known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
An effective amount for tumor therapy may be measured by its ability to stabilize disease progression and/or ameliorate symptoms in a patient, and preferably to reverse disease progression, e.g., by reducing tumor size. The ability of a therapy or composition of the present disclosure to inhibit cancer may be evaluated by in vitro assays, e.g., as described in the examples, as well as in suitable animal models that are predictive of the efficacy in human tumors. Suitable dosage regimens will be selected in order to provide an optimum therapeutic response in each particular situation, for example, administered as a single bolus or as a continuous infusion, and with possible adjustment of the dosage as indicated by the exigencies of each case.
The therapies and compositions of the present disclosure may be administered by any method for administering peptides, proteins or antibodies accepted in the art, and are typically suitable for parenteral administration. As used herein, “parenteral administration” includes any route of administration characterized by physical breaching of a tissue of a subject and administration through the breach in the tissue, thus generally resulting in the direct administration into the blood stream, into muscle, or into an internal organ. Parenteral administration thus includes, but is not limited to, administration by injection, by application through a surgical incision, by application through a tissue-penetrating non-surgical wound, and the like. In particular, parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal, intracisternal, intravenous, intraarterial, intrathecal, intraurethral, intracranial, intratumoral, and intrasynovial injection or infusions. Particular embodiments include the intravenous and the subcutaneous routes. In some embodiments, the administration is IV injection, e.g., IV infusion.
In some embodiments, the therapies and compositions of the present disclosure may be administered according to an exemplary dosing regimen described in Example 1.
For example, in certain embodiments, the anti-EGFR antibodies component or antigen-binding portion may be administered at a dose of 1 , 3, 6, 9, 12, 15, or 18 mg/kg (e.g., as a monotherapy, or as part of a combination therapy, as described herein). ). In some embodiments, the anti-EGFR antibodies component or antigen-binding portion may be administered at one of said doses as a loading dose and a different one of said doses as a maintenance dose, for example a loading dose of 9 mg/kg followed by a maintenance dose of 6 mg/kg. In particular embodiments, the anti-EGFR antibodies component or antigen-binding portion thereof is administered every 1 , 2, 3, or 4 weeks.
In particular embodiments, the anti-EGFR antibodies component or antigen-binding portion is administered first day every 1 , 2, 3, or 4, weeks. Further, 4 weeks constitutes a cycle of 28 days, the anti-EGFR antibodies component or antigen-binding portion is administered in cycle 1 , 2, 3 etc.
In certain embodiments, the trifluridine/tipiracil component may be administered at a dose of 35mg/m2 twice daily (bid) (e.g., as part of a combination therapy as described herein). In particular embodiments, the trifluridine/tipiracil component is administered during a cycle of 28 days on days 1 -5 of week 1 and days 1 -5 of week 2.
Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Throughout this specification and embodiments, the words “have” and “comprise,” or variations such as “has,” “having,” “comprises,” or “comprising,” will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
All publications and other references mentioned herein are incorporated by reference in their entirety. Although a number of documents are cited herein, this citation does not constitute an admission that any of these documents forms part of the common general knowledge in the art.
In order that the present disclosure may be better understood, the following examples are set forth. These examples are for purposes of illustration only and are not to be construed as limiting the scope of the present disclosure in any manner.
ABBREVIATIONS
Abbreviations used herein are shown below.
ADA : Anti-Drug Antibody
AE : Adverse Event
ALT : Alanine Aminotransferase
ANC : Absolute Neutrophil Count
AST : Aspartate Aminotransferase
AUC : Area Under the Plasma Concentration-Time Curve
BICR Blinded Independent Central Review
BID : Twice Daily
BOR Best Overall Response
Cmax : Maximum Plasma Concentration
CR : Complete Response
CRC : Colorectal Cancer ctDNA : Circulating tumour DNA
CT : Computerized Tomography
DCR Disease Control Rate
DLT Dose-Limiting Toxicity
DoR Duration Of Response
ECD Extracellular Domain
ECOG Eastern Cooperative Oncology Group
EGFR Epidermal Growth Factor Receptor
EMA European Medicines Agency
EORTC European Organization For Research And Treatment Of
Cancer
EOT End Of Treatment
FAS Full Analysis Set
GCP : Good Clinical Practice
GGT : Gamma-Glutamyl Transferase (Gamma-Glutamyl
Transpeptidase)
HER Human Epidermal Growth Factor Receptor
HIV Human Immunodeficiency Virus
HR Hazard Ratio
HRT Hormone Replacement Therapy
IE Intercurrent Event
IMP Investigational Medicinal Product: a pharmaceutical form of an active ingredient or placebo being tested or used as a reference in a clinical trial (i.e. futuximab/modotuximab and trifluridine/tipiracil in this study)
IV Intravenous (Route)
MA : Marketing Authorization
MMR : Mismatch Repair
MSI : Microsatellite Instability
NE Not Evaluable
OR : Objective Response ORR : Objective Response Rate
OS : Overall Survival
PD Progressive Disease
PFS Progression Free Survival
PK Pharmacokinetics
PO : Orally
PR Partial Response
PRO Participant-Reported Outcome
PS Performance Status
QoL : Quality of Life
RECIST : Response Evaluation Criteria In Solid Tumours
SAE : Serious Adverse Event
SD : Stable Disease
TEAE : Treatment Emergent Adverse Event
TESAE : Treatment Emergent Serious Adverse Events
TtPS2 : Time to ECOG Performance Status > 2 TTR : Time To Response
ULN Upper Limit of Normal VEGF : Vascular Endothelial Growth Factor
EXAMPLES
Example 1: Phase 3 clinical protocol for futuximab/modotuximab in combination therapy This example describes a clinical trial protocol for an international, randomised, open label, multi-centre, two-arm Phase 3 study to evaluate futuximab/modotuximab in combination with trifluridine/tipiracil versus trifluridine/tipiracil monotherapy in approximately 500 patients with KRAS/NRAS and BRAF WT metastatic colorectal cancer (mCRC) who were previous treated by chemotherapy (including oxaliplatin, irinotecan and 5- fluorouracil, anti-VEGF agents) and with anti-EGFR mAb therapy for more than 4 months. Maximum study duration for each participating patient: each patient will participate in the study until confirmed disease progression, loss to followup, an adverse event leading to withdrawal, significant noncompliance with the study protocol, withdrawal of consent, end of study, or death from any cause.
Study Design
A. Endpoints for the safety Lead-in part
A primary objective is to assess safety and tolerability of futuximab/modotuximab in combination with trifluridine/tipiracil. The corresponding primary endpoints are:
- incidence of dose-limiting toxicities (DLTs)
- incidence, severity and relationship of Treatment Emergent Adverse Events (TEAEs) and Treatment Emergent Serious Adverse Events (TESAEs)
- AEs leading to dose interruption, modification, delays and permanent treatment stop
- changes from baseline in key laboratory safety assessments, and vital signs
A first secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil using Response Evaluation Criteria in Solid Tumours (RECIST) in terms of: Objective Response Rate (ORR), Best Overall Response (BOR), Disease Control Rate (DCR), Progression Free Survival (PFS). The corresponding secondary endpoints are:
- Objective Response (OR): achievement of complete response (CR) or partial response (PR)
- BOR: the best response recorded from screening through disease progression/recurrence
- Disease Control (DC): achievement of best response: CR: disappearance of all target lesions, PR: > 30% decrease in the sum of the longest diameter of target lesions, or Stable Disease (SD): > 24 weeks
- PFS: the time from the first administration of first IMP date until the date of the investigator-assessed radiological disease progression or death, whichever occurs first
A second secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil in terms of Overall Survival (OS). The corresponding second secondary endpoint is :
- OS: the time from the first administration of first IMP date to death from any cause.
A third secondary objective is to characterise the pharmacokinetic (PK) profile of futuximab/modotuximab, trifluridine and tipiracil in the combination of futuximab/modotuximab with trifluridine/tipiracil. The corresponding third secondary endpoint is :
- Derived PK parameters (e.g. Cmax, AUC) for futuximab/modotuximab, trifluridine and tipiracil.
The fourth secondary objective is to evaluate the immunogenicity of futuximab/modotuximab (i.e. occurrence of anti-drug antibody [ADA]). The corresponding fourth secondary endpoint is :
- ADA development for futuximab/modotuximab.
B. Endpoints for the Phase 3 part
The primary objective is to compare OS of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in patients with tumours that are KRAS/NRAS and BRAF WT (Double negative [DN]). The corresponding primary endpoint is :
- OS (in DN) is defined as the time from date of randomisation into the study to death from any cause
The key secondary objective is to compare OS of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in patientswith tumours that are KRAS/NRAS, BRAF WT and EGFR- extracellular domain WT (Triple negative [TN]). The corresponding key secondary endpoint is :
- OS (in DN) is defined as the time from date of randomisation into the study to death from any cause.
The first secondary objective is to compare anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy per Blinded Independent Central Review (BICR) assessment using RECIST in terms of Progression Free Survival (PFS), Objective Response Rate (ORR), Disease Control Rate (DCR), Duration of response (DoR) and Time to Response (TTR). The corresponding first secondary endpoints are :
- PFS: the time from date of randomisation until the date of the centrally-assessed radiological disease progression or death, whichever occurs first
- OR: achievement of confirmed, complete response (CR) or partial response (PR)
- DC: achievement of best response: CR: disappearance of all target lesions, PR: > 30% decrease in the sum of the longest diameter of target lesions, or Stable Disease (SD): > 24 weeks
- DoR: the time from the first documentation of confirmed tumour response (CR or PR) to the first documentation of objective tumour progression or to death due to any cause
- TTR: the time from randomization until first radiologically confirmed tumour response (CR or PR)
The second secondary objective is to assess anti-tumour activity of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy in terms of Time to Next Treatment (TTNT) and Time to ECOG Performance Status > 2 (TtPS2). The corresponding second secondary endpoints are :
- TTNT: the time from the randomisation to initiation of the next systemic anti-cancer therapy - TtPS2: the time from the date of randomization to the date when ECOG PS score of > 2 is observed for the first time
The third secondary objective is to further evaluate and to compare the safety profile of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy. The corresponding third secondary endpoints are :
- Incidence, severity and relationship of TEAEs and TESAEs collected from administration of the first dose of IMPs
- AEs leading to dose interruption, dose delays and permanent treatment stop
- Changes from baseline in key laboratory safety assessments, and vital signs
The fourth secondary objective is to compare Quality of life (QoL) of futuximab/modotuximab in combination with trifluridine/tipiracil vs trifluridine/tipiracil monotherapy. The corresponding fourth secondary endpoint is :
- QoL is assessed by 2 questionnaires European Organization for Research and Treatment of Cancer (EORTC) QLQ-C30 and EQ-5D-5L
The fifth secondary objective is to characterize the pharmacokinetic (PK) profile of futuximab/modotuximab, trifluridine and tipiracil in the combination futuximab/modotuximab with trifluridine/tipiracil. The corresponding fifth secondary endpoint is :
- Derived PK parameters (e.g. Cmax, AUC) for futuximab/modotuximab, trifluridine and tipiracil
The sixth secondary objective is to further evaluate the immunogenicity of futuximab/modotuximab (/.e. occurrence of antidrug antibody [ADA]). The corresponding first sixth endpoint is :
- ADA development for futuximab/modotuximab
Methodology
The study (Figure 1 ) will comprise two parts a Safety Lead-In part (Figure 2) in approximately 25 patientsand a randomised Phase 3 part (Figure 3) in approximately 500 patients.
The Safety Lead-In part is an open-label, single-arm and non-random ised study phase in approximately 25 patients. During this part the safety and tolerability of futuximab/modotuximab in combination with trifluridine/tipiracil will be evaluated prior to the start of the Phase 3 part. Efficacy parameters, futuximab/modotuximab and trifluridine/tipiracil PK and futuximab/modotuximab immunogenicity will also be evaluated as secondary endpoints.
The therapeutic regimen for the Safety Lead-In part is as follows:
- trifluridine/tipiracil (35 mg/m2 BID) 5 days on/2 days off over 2 weeks followed by 14 days off, before futuximab/modotuximab administration.
- futuximab/modotuximab (9 mg/kg) loading dose on C1 D1 followed by (6 mg/kg) weekly maintenance doses.
The Phase 3 part is of open-label, multi-centre, parallel-group, 2-arm, randomised design. Approximately 500 patients previously treated by chemotherapy (including oxaliplatin, irinotecan and 5-fluorouracil, anti-VEGF agents) and who have received previous treatment with anti-EGFR mAb therapy for > 4 months, KRAS/NRAS and BRAF WT mCRC will be randomised in a 1 :1 ratio to either futuximab/modotuximab in combination with trifluridine/tipiracil (Arm A) or trifluridine/tipiracil (Arm B), and stratified according to performance status (PS) (0 versus 1 ), previous regimens of treatment (2 versus > 3) and by the presence or absence of EGFR ECD mutations.
Futuximab/modotuximab and trifluridine/tipiracil are the IMPs. Table 2 provides a description of the IMPs. Table 2
Figure imgf000056_0001
USP — United States Pharmacopeia
Screening criteria
The medical and therapeutic criteria include :
- patient being male or female > 18 years old;
- patients must have histologically or cytologically confirmed adenocarcinoma of mCRC, not amenable to surgical intervention due to either medical ontraindications or non-resectability of the tumour;
- patients should be without RAS (KRAS and NRAS) mutations in any of the following codons: exon 2: codon 12, 13;exon 3: codon 59, 61 ; exon 4: codon 117, 146 and without BRAF V600E mutation based on ctDNA screening blood test analysis patients;
- patients must have measurable or non-measurable lesion according to RECIST;
- Patientsmust have received at least 2 prior regimens of standard chemotherapy for mCRC and had demonstrated progressive disease or intolerance to their last regimen. The following characteristics apply:
Prior standard chemotherapy must not have included trifluridine/tipiracil but must have included all of the following agents approved and available in each country: fluoropyrimidines, irinotecan and oxaliplatin; at least one anti-VEGF pathway inhibitor (bevacizumab and/or aflibercept and/or ramucirumab and/or regorafenib); at least one anti-EGFR mAb (cetuximab or panitumumab).
Patients must have progressed based on clinical evaluation during or within 6 months of the last administration of the last standard chemotherapy regimen. Patients who have withdrawn from standard treatment due to unacceptable toxicity warranting discontinuation of treatment and precluding retreatment with the same agent prior to progression of disease will be eligible to enter the study.
Patients who received adjuvant/neoadjuvant chemotherapy and had recurrence during or within 6 months of completion of the adjuvant/neoadjuvant chemotherapy are permitted to count the adjuvant/neoadjuvant therapy as one regimen of chemotherapy.
- Patients should have received previous treatment with commercially available anti-EGFR mAbs for > 4 months.
- Ability to swallow oral medication.
- Estimated life expectancy > 12 weeks. This criterion should be rechecked at inclusion visit.
- ECOG performance status 0 or 1 (or equivalent Karnofsky PS of 70% to 100%).
The inclusion criteria include:
- adequate haematological function based on the last assessment performed within 7 days prior to the first IMP administration, defined as: (1 ) absolute neutrophil count (ANC) > 1.5 x 109/L; (2) haemoglobin > 90 g/L; (3) platelet count > 100 x 109/L; and (4) adequate coagulation function for all patients.
- adequate renal function based on the last assessment performed within 7 days prior to the first IMP administration defined as creatinine clearance > 50 mL/min assessed using the Cockcroft & Gault formula.
- adequate hepatic function based on the last assessment performed within 7 days prior to the first IMP administration, defined as: (1 ) total serum bilirubin < 1 .5 x upper limit of normal (ULN); (2) aspartate aminotransferase (AST; SGOT) and alanine aminotransferase (ALT; SGPT) < 2.5 x ULN (if liver function abnormalities are due to underlying liver metastasis, AST [SGOT] and ALT [SGPT] < 5 x ULN).
- Serum potassium, serum phosphates, serum magnesium within normal limits with or without supplementation based on the last assessment performed within 7 days prior to the first IMP administration.
- Women of childbearing potential (WOCBP) must use a highly effective method of birth control during study treatment beginning within 2 weeks prior to the first IMP dose and continuing at least 6 months after the last dose of IMP.
- Male patients with WOCBP partners must use a condom during the study and until at least 6 months after the last dose of IMP.
Criteria for Evaluation
Tumour assessments/imaging of the chest, abdomen, and pelvis at a minimum will be obtained at each time point listed below for all patients:
Baseline: tumour assessment will be done within 28 days prior to the first administration of first IMP.
Treatment period: tumour assessments will be done every 8 weeks from C1 D1 (± 7 calendar days) until radiologic progression is documented.
Withdrawal visit: tumour assessments will be done only if not performed within previous 8 weeks. Every effort should be made to perform the EOT tumour assessments prior to the start of new anti-cancer therapy.
Follow-up period: tumour assessments will be done for patients who were withdrawn for reasons other than radiologic disease progression or consent withdrawal, every 8 weeks during the follow up period until the participant experienced radiologic progression, regardless of the initiation of a new anticancer therapy.
Tumour assessments will be performed as per RECIST version 1.1 at baseline and then every 8 weeks from C1 D1 (± 7 calendar days) until radiologic progression, death or end of study, whichever occurs first. The definition of responses for Target and Non-target lesions is presented below:
Target lesions
Compete Response (CR): the disappearance of all target lesions. Any pathological lymph nodes must have reduction in short axis to < 10 mm.
Partial Response (PR): at least a 30% decrease in the sum of diameters of the target lesions, taking as a reference the baseline sum diameters.
Progressive Disease (PD): at least a 20% increase in the sum of diameters of the target lesions, taking as a reference the smallest sum on study, including the baseline sum. In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. Definitive new lesion presence also indicates progression.
Stable Disease (SD): neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as a reference the smallest sum diameter while on study.
Non-target lesions
Compete Response (CR): The disappearance of all non-target lesions and normalisation of tumour marker level. All lymph nodes must be non- pathological morphologically (/.e., < 10 mm in short axis in size).
Non-CR/Non-PD: A persistence of > 1 non-target lesion(s) and/or maintenance of tumour marker level above the normal limits (not reaching the extent of ‘unequivocal progression).
Progressive Disease (PD): Unequivocal progression of existing non-target lesions
The investigator should consider all lesions (target and non-target) in assessing the tumor burden at repeat imaging prior to decide whether to continue treatment. When the participant has measurable disease and non- measurable disease, to achieve “unequivocal progression” on the basis of the non-target disease, there must be an overall level of substantial worsening in non-target disease such that, even in presence of SD or PR in target disease, the overall tumour burden has increased sufficiently to merit discontinuation of therapy. A modest ‘increase’ in the size of one or more non-target lesions is usually not sufficient to quality for unequivocal progression status.
Figure imgf000061_0001
Table 3:
Figure imgf000061_0003
Figure imgf000061_0002
Figure imgf000062_0001
Figure imgf000062_0002
Figure imgf000063_0001
Figure imgf000063_0002

Claims

63
CLAIMS A combination comprising a) an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof that competes or cross-com petes for binding to human EGFR with, or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component. The combination of claim 1 wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H- CDR) 1 -3 and light chain complementarity-determining regions (L- CDR) 1 -3 that comprise the amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively. The combination of claim 2, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively. The combination of claim 3, wherein the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or 64 b) SEQ ID NOs: 19 and 20, respectively. The combination of claims 1-4, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H- CDR) 1 -3 and light chain complementarity-determining regions (L- CDR) 1 -3 that comprise the amino acid sequences of SEQ ID NOs: 1 - 6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with H-CDR1 -3 and L-CDR1 -3 that comprise the amino acid sequences of SEQ ID NOs: 11-16, respectively. The combination of claim 5, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with a VH and VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively. The combination of claim 6, wherein the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively. A method of enhancing immunity in a human patient in need thereof, comprising administering to the patient a) an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof that competes or cross-com petes for binding to human EGFR with, 65 or binds to the same epitope of human EGFR as, an antibody that comprises the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component. The method of claim 8, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L-CDR) 1-3 that comprise the amino acid sequences of: a) SEQ ID NOs: 1 -6, respectively; or b) SEQ ID NOs: 11 -16, respectively. The method of claim 9, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of: a) SEQ ID NOs: 7 and 8, respectively; or b) SEQ ID NOs: 17 and 18, respectively. The method of claim 10, wherein the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of: a) SEQ ID NOs: 9 and 10, respectively; or b) SEQ ID NOs: 19 and 20, respectively. The method of any one of claims 8-11 , wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with heavy chain complementarity-determining regions (H-CDR) 1 -3 and light chain complementarity-determining regions (L- 66
CDR) 1 -3 that comprise the amino acid sequences of SEQ ID NOs: 1 - 6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with H-CDR1 -3 and L-CDR1 -3 that comprise the amino acid sequences of SEQ ID NOs: 11-16, respectively. The method of claim 12, wherein the anti-EGFR component comprises an anti-EGFR antibody or an antigen-binding portion thereof with a heavy chain variable domain (VH) and light chain variable domain (VL) that comprise the amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof with a VH and VL that comprise the amino acid sequences of SEQ ID NOs: 17 and 18, respectively. The method of claim 13, wherein the anti-EGFR component comprises an anti-EGFR antibody with a heavy chain (HC) and light chain (LC) that comprise the amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody with an HC and LC that comprise the amino acid sequences of SEQ ID NOs: 19 and 20, respectively. The method of any one of claims 8-14, wherein the anti-EGFR component is futuximab, modotuximab, or futuximab + modotuximab. The method of claim 8, comprising administering to the patient: a) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 - 3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 1 -6, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the H-CDR1 -3 and L-CDR1 -3 amino acid sequences of SEQ ID NOs: 11-16, respectively; and a trifluridine/tipiracil component ; 67 b) an anti-EGFR component comprising an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 7 and 8, respectively, and an anti-EGFR antibody or an antigen-binding portion thereof comprising the VH and VL amino acid sequences of SEQ ID NOs: 17 and 18, respectively; and a trifluridine/tipiracil component ; or c) an anti-EGFR component comprising an anti-EGFR antibody comprising the HC and LC amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and an anti-EGFR antibody comprising the HC and LC amino acid sequences of SEQ ID NOs: 19 and 20, respectively; and a trifluridine/tipiracil component.
17. The method of any one of claims 8-16, wherein the one or two anti- EGFR antibodies or antigen-binding portions thereof and the trifluridine/tipiracil component are administered to the patient concurrently.
18. The method of any one of claims 8-16, wherein the one or two anti- EGFR antibodies or antigen-binding portions thereof and the trifluridine/tipiracil component are administered to the patient sequentially.
19. The method of any one of claims 8-18, wherein the patient has cancer.
20. The method of claim 19, wherein the cancer is a hematological malignancy.
21 . The method of claim 19, wherein the cancer is a solid tumor. The method of claim 21 , wherein the cancer is colorectal cancer or gastric cancer. The method of claim 22, wherein the cancer is metastatic colorectal cancer (mCRC). The method of any one of claims 8-23, wherein the anti-EGFR component is administered at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, optionally wherein the component is administered weekly. The method of any one of claims 8-23, wherein the trif lurid ine/tipiracil component is administered at a dose of 20 to 80 mg/m2/day, optionally wherein the component trifluridine/tipiracil is divided in two portions per day for five consecutive days and then a 2-days rest period. The method of claim 25, wherein the trifluridine/tipiracil component is administered at a dose of 20mg/m2, 25mg/m2, 30mg/m2, 35mg/m2. The method of any one of claims 8-26, wherein the one or two anti- EGFR antibodies or antigen-binding portions thereof are formulated for intravenous infusion. The method of any one of claims 8-26, wherein the trifluridine/tipiracil component is formulated for orally administration. A method of treating an advanced solid tumor malignancy in a patient, comprising administering to the patient an anti-EGFR antibody component comprising one or two anti-EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and a trifluridine/tipiracil component, wherein the anti-EGFR component is administered by IV infusion at a dose of 6 mg/kg, 9 mg/kg, or a loading dose of 9 mg/kg followed by 6 mg/kg, wherein the trifluridine/tipiracil component is administered orally at a dose of 35mg/m2. A method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti- EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; optionally wherein the cancer is locally advanced and/or unresectable, and optionally wherein the patient has failed on first-line standard of care therapy. The method of claim 30, wherein
(a) the anti-EGFR antibody component is administered at a loading dose dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , day 8, day 15, day 22 of each cycle, and
(b) the trifluridine/tipiracil component is administered at a dose of 35mg/m2 on days 1 to day 5 and days 8 to day 12 of each cycle, wherein the anti-EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally. A method of treating metastatic colorectal cancer in a patient, comprising administering to the patient: a) an anti-EGFR antibody component comprising one or two anti- EGFR antibodies comprising the heavy and light chain amino acid sequences of SEQ ID NOs: 9 and 10, respectively, and/or SEQ ID NOs: 19 and 20, respectively; and b) a trifluridine/tipiracil component; wherein the patient is (i) without RAS mutations in any of the following codons: codons 12 and 13 in exon 2, codons 59 and 61 in exon 3, and codons 117 and 146 in exon 4; and/or (ii) without the BRAF V600E mutation. The method of claim 32, wherein
(a) the anti-EGFR antibody component is administered at a loading dose dose of 9mg/kg on Cycle 1 Day 1 (C1 D1 ) once weekly on day 1 , day 8, day 15, day 22 of each cycle, and
(b) the trifluridine/tipiracil component is administered at a dose of 35mg/m2 on days 1 to day 5 and days 8 to day 12 of each cycle, wherein the anti-EGFR antibody component is administered via IV infusion and the trifluridine/tipiracil component is administered orally. The method of any one of claims 8-33, further comprising administering to the patient radiation therapy, or at least one of a chemotherapeutic agent, an anti-neoplastic agent, and an anti- angiogenic agent. The method of any one of claims 16-34, wherein the treatment results in one or more of the following: a) improved objective response rate; b) improved clinical benefit rate; c) improved duration of response; d) increased progression-free survival; and e) increased overall survival. A pharmaceutical composition comprising an anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen- 71 binding portions thereof as defined in any one of claims 1 -7, and further comprising a trifluridine/tipiracil component, and a pharmaceutically acceptable excipient.
37. A pharmaceutical composition comprising the components of the method of any one of claims 8-35.
38. A pharmaceutical composition of any one of claims 36-37 for use in treating a human patient in a method of any one of claims 8-35.
39. An anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as defined in any one of claims 1 -7 for use in enhancing immunity in a human patient in need thereof in combination with a trifluridine/tipiracil component.
40. An anti-EGFR antibody component comprising one or two anti-EGFR antibodies or antigen-binding portions thereof as defined in any one of claims 1 -7 in combination with a trifluridine/tipiracil component for use in treating a human patient in a method of any one of claims 8-35.
41 . Use of an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as defined in any one of claims 1 -7 in combination with a trifluridine/tipiracil component for the manufacture of a medicament for enhancing immunity in a human patient in need thereof.
42. Use of an anti-EGFR antibody component comprising one or two anti- EGFR antibodies or antigen-binding portions thereof as defined in any one of claims 1 -7 in combination with a trifluridine/tipiracil component for the manufacture of a medicament for treating a human patient in a method of any one of claims 8-35. 72 The combination of claims 1 to 7 for use in a method of treatment of cancer in a subject that has been subjected to a prior treatment regimen.
PCT/EP2022/085198 2021-12-10 2022-12-09 Cancer therapy targeting egfr WO2023105051A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21306751 2021-12-10
EP21306751.5 2021-12-10

Publications (1)

Publication Number Publication Date
WO2023105051A1 true WO2023105051A1 (en) 2023-06-15

Family

ID=80035092

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/085198 WO2023105051A1 (en) 2021-12-10 2022-12-09 Cancer therapy targeting egfr

Country Status (3)

Country Link
AR (1) AR127893A1 (en)
TW (1) TW202339796A (en)
WO (1) WO2023105051A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0763529A1 (en) 1995-03-29 1997-03-19 Taiho Pharmaceutical Company Limited Uracil derivatives, and antitumor effect potentiator and antitumor agent containing the same
WO2000009560A2 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
WO2008104183A2 (en) 2007-03-01 2008-09-04 Symphogen A/S Recombinant anti-epidermal growth factor receptor antibody compositions
WO2012125864A2 (en) * 2011-03-15 2012-09-20 Merrimack Pharmaceuticals, Inc. Overcoming resistance to erbb pathway inhibitors
EP3042669A1 (en) * 2013-09-06 2016-07-13 Taiho Pharmaceutical Co., Ltd. Antitumor agent and antitumor effect enhancer
WO2019043059A1 (en) * 2017-08-30 2019-03-07 Symphogen A/S Compositions and methods for treating cancer with anti-egfr antibodies

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0763529A1 (en) 1995-03-29 1997-03-19 Taiho Pharmaceutical Company Limited Uracil derivatives, and antitumor effect potentiator and antitumor agent containing the same
WO2000009560A2 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
WO2008104183A2 (en) 2007-03-01 2008-09-04 Symphogen A/S Recombinant anti-epidermal growth factor receptor antibody compositions
US8414896B2 (en) * 2007-03-01 2013-04-09 Symphogen A/S Recombinant anti-epidermal growth factor receptor antibody compositions
WO2012125864A2 (en) * 2011-03-15 2012-09-20 Merrimack Pharmaceuticals, Inc. Overcoming resistance to erbb pathway inhibitors
EP3042669A1 (en) * 2013-09-06 2016-07-13 Taiho Pharmaceutical Co., Ltd. Antitumor agent and antitumor effect enhancer
WO2019043059A1 (en) * 2017-08-30 2019-03-07 Symphogen A/S Compositions and methods for treating cancer with anti-egfr antibodies

Non-Patent Citations (35)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING COMPANY
BASELGA ET AL., PHARMAC. THERAPEUT, vol. 64, 1994, pages 127 - 154
BASELGA ET AL., PHARMAC. THERAPEUT., vol. 64, 1994, pages 127 - 154
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 6
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 878 - 83
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 17
HOLLIGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 8
HONEGGERPLUCKTHUN, J. MOL. BIOL., vol. 309, no. 3, 2001, pages 657 - 70
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 83
INVESTIGATIONAL NEW DRUGS, vol. 26, no. 5, 2008, pages 445 - 454
IRENE V. BIJNSDORP ET AL: "Molecular mechanism underlying the synergistic interaction between trifluorothymidine and the epidermal growth factor receptor inhibitor erlotinib in human colorectal cancer cell lines", CANCER SCIENCE, vol. 101, no. 2, 1 February 2010 (2010-02-01), JP, pages 440 - 447, XP055322963, ISSN: 1347-9032, DOI: 10.1111/j.1349-7006.2009.01375.x *
KARNES ET AL., GASTROENTEROLOGY, vol. 114, 1998, pages 930 - 939
KATO TAKESHI ET AL: "Safety and efficacy of panitumumab in combination with trifluridine/tipiracil for pre-treated patients with unresectable, metastatic colorectal cancer with wild-type RAS: The phase 1/2 APOLLON study", INTERNATIONAL JOURNAL OF CLINICAL ONCOLOGY, SPRINGER SINGAPORE, SINGAPORE, vol. 26, no. 7, 29 April 2021 (2021-04-29), pages 1238 - 1247, XP037485319, ISSN: 1341-9625, [retrieved on 20210429], DOI: 10.1007/S10147-021-01902-2 *
KIPRIYANOV ET AL., HUMAN ANTIBODIES AND HYBRIDOMAS, vol. 6, 1995, pages 93 - 101
KIPRIYANOV ET AL., MOL. IMMUNOL., vol. 31, 1994, pages 1047 - 58
LEFRANC ET AL., DEV COMP IMMUNOL, vol. 27, no. 1, 2003, pages 55 - 77
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 45
MARTIN ET AL., EMBO J., vol. 13, 1994, pages 5303 - 9
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 4
MODJTAHEDI ET AL., INT. J. ONCOLOGY., vol. 4, 1994, pages 277 - 296
PEARSON, J. MOL. BIOL., vol. 276, 1998, pages 71 - 84
PEARSON, METHODS ENZYMOL., vol. 183, 1990, pages 63 - 98
PEARSON, METHODS ENZYMOL., vol. 266, 1996, pages 227 - 58
PEARSON, METHODS MOL. BIOL., vol. 132, 2000, pages 185 - 219
PRICE ET AL., EUR. J. CANCER, vol. 32A, 1996, pages 1977 - 1982
PROTEIN ENG., vol. 10, 1997, pages 949 - 57
QURESHI SADAF ET AL: "Current Treatment Landscape for Third- or Later-Line Therapy in Metastatic Colorectal Cancer", CURRENT COLORECTAL CANCER REPORTS, SPRINGER US, BOSTON, vol. 17, no. 6, 23 October 2021 (2021-10-23), pages 131 - 141, XP037647321, ISSN: 1556-3790, [retrieved on 20211023], DOI: 10.1007/S11888-021-00469-4 *
SALOMON DS ET AL., CRITICAL REVIEWS IN ONCOLOGY/HAEMATOLOGY, vol. 19, 1995, pages 183 - 232
SALOMON ET AL., CRIT. REV. ONCOLOGY/HAEMATOLOGY, vol. 19, 1995, pages 183 - 232
TABERNERO J ET AL: "478O Efficacy and safety of Sym004 in refractory metastatic colorectal cancer with acquired resistance to anti-EGFR therapy: Results of a randomized phase II study (RP2S)", 30 September 2017 (2017-09-30), pages 1 - 1, XP055919306, Retrieved from the Internet <URL:https://www.annalsofoncology.org/action/showPdf?pii=S0923-7534%2820%2938965-1> [retrieved on 20220510] *
TRAUNECKER ET AL., EMBO J., 1991, pages 3655 - 9
TRAUNECKER ET AL., INT. J. CANCER, vol. 7, 1992, pages 51 - 2
VAN CUTSEM E. ET AL: "ESMO consensus guidelines for the management of patients with metastatic colorectal cancer", ANNALS OF ONCOLOGY, vol. 27, no. 8, 1 August 2016 (2016-08-01), NL, pages 1386 - 1422, XP055917783, ISSN: 0923-7534, DOI: 10.1093/annonc/mdw235 *
WOODBURN ET AL., PHARMACOL. THERAP., vol. 82, 1999, pages 241 - 250
WU ET AL., J. CLIN. INVEST., vol. 95, 1995, pages 1897 - 1905

Also Published As

Publication number Publication date
TW202339796A (en) 2023-10-16
AR127893A1 (en) 2024-03-06

Similar Documents

Publication Publication Date Title
EP3334431B1 (en) 5-bromo-2,6-di-(1h-pyrazol-l-yl)pyrimidin-4-amine for use in the treatment of cancer
JP7137563B2 (en) Anti-Siglec-7 antibody for cancer therapy
JP6967853B2 (en) Antibodies that bind to ErbB-2 and ErbB-3
JP2024023222A (en) Combination therapies targeting pd-1, tim-3, and lag-3
JP2024054226A (en) Treatment of resistant cancers by administration of anti-HER2 antibody-drug conjugates
JP2008508858A (en) Inhibition of macrophage-stimulated protein receptor (RON)
US11634500B2 (en) Anti-CD73 antibodies and compositions
JP2022533418A (en) Anti-ROR1/anti-CD3 bispecific binding molecules
KR20220139916A (en) Anti-ROR1 Antibodies and Compositions
WO2022100590A1 (en) Adcc-enhanced humanized antibody for claudin 18a2 and application thereof
WO2021115303A1 (en) Anti-claudin18.2 monoclonal antibody, preparation method therefor and use thereof
US20210030888A1 (en) Anti-tissue factor antibody-drug conjugates and their use in the treatment of cancer
WO2021058735A1 (en) Cancer treatment with anti-met antibody compositions
JP2023510132A (en) Methods of treating cancer using combinations of PD-1 antagonists, ILT4 antagonists and chemotherapeutic agents
CN103417965B (en) A kind of pharmaceutical composition containing anti-VEGF antibody
WO2023105051A1 (en) Cancer therapy targeting egfr
JP2023522630A (en) Anti-FLT3 antibodies and compositions
EP3164154A1 (en) Combination therapy
BR112019020507A2 (en) erbb-2 targeting agent and a bispecific antibody with antigen binding sites that bind an epitope to an extracellular part of erbb-2 and erbb-3 for treating an individual with an erbb-2, erbb-2 / positive tumor erbb-3
WO2019072566A1 (en) Combination of anti-il10 and anti-pd1 antibodies in cancer treatment
TW202323298A (en) Cancer therapy targeting nkg2a
JP6964113B2 (en) 5-Bromo-2,6-di- (1H-pyrazole-1-yl) pyrimidine-4-amine for use in the treatment of cancer
JP2024520577A (en) Anti-NKG2A antibodies and compositions
TW202415406A (en) Methods of treating cancer and the pharmaceutical compositions thereof

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: 22835619

Country of ref document: EP

Kind code of ref document: A1