WO2024095173A1 - Methods of treating cancers - Google Patents

Methods of treating cancers Download PDF

Info

Publication number
WO2024095173A1
WO2024095173A1 PCT/IB2023/060998 IB2023060998W WO2024095173A1 WO 2024095173 A1 WO2024095173 A1 WO 2024095173A1 IB 2023060998 W IB2023060998 W IB 2023060998W WO 2024095173 A1 WO2024095173 A1 WO 2024095173A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
bcma
subject
bispecific antibody
dose
Prior art date
Application number
PCT/IB2023/060998
Other languages
French (fr)
Inventor
Jenna GOLDBERG
Arnob BANERJEE
Rachel KOBOS
Original Assignee
Janssen Biotech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Biotech, Inc. filed Critical Janssen Biotech, Inc.
Publication of WO2024095173A1 publication Critical patent/WO2024095173A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • 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
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin

Definitions

  • BCMA x CD3 bispecific antibodies Disclosed are methods of treating cancers with BCMA x CD3 bispecific antibodies and combinations comprising BCMA x CD3 bispecific antibodies, anti-CD38 antibodies, and immunomodulatory drugs.
  • M-proteins monoclonal proteins
  • pathological immunoglobulins or fragments of such which have lost their function.
  • M-proteins monoclonal proteins
  • the proliferation of multiple myeloma cells leads to subsequent displacement from the normal bone marrow niche, while overproduction of M-proteins causes characteristic osteolytic lesions, increased susceptibility to infections, hypercalcemia, renal insufficiency or failure, and neurological complications.
  • Treatment options for multiple myeloma have improved over time and vary depending on the aggressiveness of the disease, underlying prognostic factors, physical condition of the patient, and existing comorbidities.
  • Therapeutic options include proteasome inhibitors (Pls), immunomodulatory drugs (IMiDs), alkylating agents, monoclonal antibodies (mAbs), antibody drug conjugate, histone deacetylase inhibitor, nuclear protein export inhibitor, chimeric antigen receptor (CAR) T cell therapy and stem cell transplantation.
  • proteasome inhibitors Pls
  • IMDs immunomodulatory drugs
  • mAbs monoclonal antibodies
  • CAR chimeric antigen receptor
  • a method of treating cancer in a subject in need thereof comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD3 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • IMD immunomodulatory drug
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
  • VH heavy chain variable region
  • the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
  • the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen-binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • HC1 is associated with LC1 to form a first antigen-binding site that immunospecifically binds BCMA
  • HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • the BCMA x CD3 bispecific antibody is teclistamab.
  • the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
  • the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
  • the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
  • the anti-CD38 antibody is daratumumab.
  • the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
  • the IMiD is lenalidomide.
  • the method further comprises administering a pre-treatment to the subject.
  • the pre-treatment comprises administration of a glucocorticoid, an antihistamine, an antipyretic, or a combination thereof.
  • a method of treating cancer in a subject in need thereof comprising i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of about 60 pg/kg, 240 pg/kg or 300 pg/kg or of about 3 mg, 4 mg, 15 mg, 24 mg or 25 mg; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg or 6000 pg/kg or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1200 mg to about 2400 mg; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMi
  • IMi immunomodulatory drug
  • a method of treating cancer in a subject in need thereof comprising i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of 60 pg/kg, 240 pg/kg, or 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg or 25 mg, or any combination thereof at a frequency selected from, daily, every other day, or weekly; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii
  • a method of treating cancer in a subject in need thereof comprising i) determining the subjects weight and assigning a BCMA x CD3 bispecific antibody treatment tier based on said weight, wherein if the subject’s weight is less than or equal to a pre-determined threshold the subject is in tier 1, and wherein if the subject’s weight is greater than a pre-determined threshold the subject is in tier 2; ii) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody; iii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody; iv) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody; and v) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD); wherein the BCMA x CD3 bispecific antibody,
  • the predetermined weight threshold is selected from the group consisting of 50 kg, 55 kg, 60 kg, 65 kg, or 70 kg.
  • a method of treating cancer in a subject in need thereof comprising i) administering subcutaneously to the subject one or more step-up doses of teclistamab at 60 pg/kg, 240 pg/kg, 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof, on days 2 and 4 of the start of treatment; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of teclistamab of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, 450 mg or any combination thereof, at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of
  • a method of treating cancer in a subject in need thereof comprising i) administering subcutaneously to the subject a first step-up dose of teclistamab of 60 pg/kg, on day 2 of a treatment regimen, and administering subcutaneously to the subject a second step-up dose of 240 pg/kg or 300 pg/kg on day 4 of a treatment regimen; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising a) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg, on days 8, 15, and 22 of the first treatment cycle; b) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg weekly for each treatment cycle thereafter, wherein wherein the 720 pg/
  • a method of treating cancer in a subject in need thereof comprising i) administering subcutaneously to the subject a first step-up dose of teclistamab on day 2 of a first treatment cycle and a second step-up dose of teclistamab on day 4 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the first step-up dose is 3 mg teclistamab and the second step-up dose is 15 mg teclistamab, and wherein if the subject weight greater than 60 kg then the first step-up dose is 4 mg teclistamab and the second step-up dose is 24 mg or 25 mg teclistamab; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising a) administering subcutaneously to the subject a treatment dose of teclistamab, on days 8, 15, and 22 of the first treatment
  • the methods provided for herein further comprise administering a pre-treatment regimen.
  • the pre-treatment regimen comprises i) intravenously or orally administering a therapeutically effective amount of dexamethasone, wherein the dexamethasone is administered at 20 mg on Day 1 of a first treatment cycle, 16 mg on days 2, 4, 8, 15, and 22 of a first treatment cycle, and 20 mg or 40 mg weekly during each of a second, third, and fourth treatment cycles; ii) intravenously or orally administering a therapeutically effective amount of diphenhydramine, wherein the diphenhydramine is administered at 25 mg to 50 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab; and iii) intravenously or orally administering a therapeutically effective amount of acetaminophen, wherein the acetaminophen is administered at 650 mg to 1000 mg for all doses of daratumumab and all
  • FIG. 1 is a schematic overview of the Phase 1 study of teclistamab administered in combination with daratumumab and lenalidomide, as of Nov. 2022 (MajesTEC-2 Regimen E; NCT04722146).
  • FIG. 2 illustrates the interleukin 6 profile for cohorts treated with teclistamab, daratumumab, and lenalidomide, as of Nov. 2022 (MajesTEC-2 Regimen E; NCT04722146).
  • FIG. 3 is a schematic overview of the initial Phase 3 study design for teclistamab administered in combination with daratumumab and lenalidomide (teclistamab-DR) as compared to daratumumab and lenalidomide in the absence of teclistamab, as of Nov. 2022 (MajesTEC-7; NCT05552222).
  • FIG. 4 is a schematic overview of the updated Phase 3 study design that includes 3 arms, Teclistamab-DR, Talquetamab-DR and DRd, as of Oct. 2023 (MajesTEC-7; NCT05552222).
  • any numerical value such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term “about.”
  • a numerical value typically includes ⁇ 10% of the recited value.
  • a dosage of 10 mg includes 9 mg to 11 mg.
  • the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.
  • Antibodies is meant in a broad sense and includes immunoglobulin molecules including monoclonal antibodies including murine, human, humanized and chimeric monoclonal antibodies, antigen binding fragments, multispecific antibodies, such as bispecific, trispecific, tetraspecific etc., dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding site of the required specificity.
  • “Full length antibodies” are comprised of two heavy chains (HC) and two light chains (LC) inter-connected by disulfide bonds as well as multimers thereof (e.g., IgM).
  • Each heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region (comprised of domains CHI, hinge, CH2 and CH3).
  • Each light chain is comprised of a light chain variable region (VL) and a light chain constant region (CL).
  • the VH and the VL regions may be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with framework regions (ER).
  • CDR complementarity determining regions
  • ER framework regions
  • Each VH and VL is composed of three CDRs and four FR segments, arranged from amino-to-carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • Immunoglobulins may be assigned to five major classes, IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence.
  • IgA and IgG are further sub-classified as the isotypes IgAl, IgA2, IgGl, IgG2, IgG3 and IgG4.
  • Antibody light chains of any vertebrate species may be assigned to one of two clearly distinct types, namely kappa (K) and lambda ( ), based on the amino acid sequences of their constant domains.
  • Antigen binding fragment or “antigen binding domain” refers to a portion of an immunoglobulin molecule that binds an antigen.
  • Antigen binding fragments may be synthetic, enzymatically obtainable or genetically engineered polypeptides and include the VH, the VL, the VH and the VL, Fab, F(ab')2, Fd and Fv fragments, domain antibodies (dAb) consisting of one VH domain or one VL domain, shark variable IgNAR domains, camelized VH domains, minimal recognition units consisting of the amino acid residues that mimic the CDRs of an antibody, such as FR3-CDR3-FR4 portions, the HCDR1, the HCDR2 and/or the HCDR3 and the LCDR1, the LCDR2 and/or the LCDR3.
  • VH and VL domains may be linked together via a synthetic linker to form various types of single chain antibody designs where the VH/VL domains may pair intramolecularly, or intermolecularly in those cases when the VH and VL domains are expressed by separate single chain antibody constructs, to form a monovalent antigen binding site, such as single chain Fv (scFv) or diabody; described for example in Int. Patent Publ. Nos.
  • scFv single chain Fv
  • diabody described for example in Int. Patent Publ. Nos.
  • BCMA refers to human B-cell maturation antigen, also known as CD269 or TNFRSF17.
  • a human BCMA (UniProt Q02223) contains the amino acid sequence of SEQ ID NO: 1.
  • the extracellular domain of the human BCMA encompasses residues 1-54 of SEQ ID NO: 1.
  • Bispecific refers to an antibody that specifically binds two distinct antigens or two distinct epitopes within the same antigen.
  • the bispecific antibody may have cross-reactivity to other related antigens, for example to the same antigen from other species (homologs), such as human or monkey, for example Macaca cynomolgus (cynomolgus, cyno) or Pan troglodytes, or may bind an epitope that is shared between two or more distinct antigens.
  • Cancer refers to a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth results in the formation of malignant tumors that invade neighboring tissues and may also metastasize to distant parts of the body through the lymphatic system or bloodstream.
  • a “cancer” or “cancer tissue” can include a tumor.
  • CD3 refers to a human antigen which is expressed on T cells as part of the multimolecular T cell receptor (TCR) complex and which consists of a homodimer or heterodimer formed from the association of two or four receptor chains: CD3 epsilon, CD3 delta, CD3 zeta and CD3 gamma.
  • Human CD3 epsilon comprises the amino acid sequence of SEQ ID NO: 2.
  • SEQ ID NO: 3 shows the extracellular domain of CD3 epsilon.
  • CD38 refers to the CD38 protein (synonyms: ADP-ribosyl cyclase 1, cADPr hydrolase 1, cyclic ADP-ribose hydrolase 1).
  • a human CD38 (UniProt accession no. P28907) has the amino acid sequence as shown in SEQ ID NO: 4.
  • CD38 is a single pass type II transmembrane protein with amino acid residues 1-21 representing the cytosolic domain, amino acid residues 22- 42 representing the transmembrane domain, and residues 43-300 representing the extracellular domain.
  • CH3 region or “CH3 domain” refers to the CH3 region of an immunoglobulin.
  • the CH3 region of human IgGl antibody corresponds to amino acid residues 341-446.
  • the CH3 region may also be any of the other antibody isotypes as described herein.
  • CAR Chimeric antigen receptor
  • CAR refers to engineered T cell receptors which graft a ligand or antigen specificity onto T cells (for example naive T cells central memory T cells effector memory T cells or combinations thereof).
  • CARs are also known as artificial T- cell receptors, chimeric T-cell receptors or chimeric immunoreceptors.
  • CARs comprise an extracellular domain capable of binding to an antigen, a transmembrane domain and at least one intracellular domain.
  • CAR intracellular domain comprises a polypeptide known to function as a domain that transmits a signal to cause activation or inhibition of a biological process in a cell.
  • the transmembrane domain comprises any peptide or polypeptide known to span the cell membrane and that can function to link the extracellular and signaling domains.
  • a chimeric antigen receptor may optionally comprise a hinge domain which serves as a linker between the extracellular and transmembrane domains.
  • Combination means that two or more therapeutics are administered to a subject together in a mixture, concurrently as single agents or sequentially as single agents in any order.
  • a “combination therapy” may refer to a therapeutically effective regimen that comprises administration of two or more anti-multiple myeloma agents to a subject to treat multiple myeloma.
  • CDR complementarity determining regions
  • CDR CDR
  • HCDR1 CDR1
  • HCDR2 CDR3
  • LCDR1 CDR2
  • LCDR3 CDR3
  • “Enhance” or “enhanced” refers to enhancement in one or more functions of a test molecule when compared to a control molecule or a combination of test molecules when compared to one or more control molecules.
  • Exemplary functions that can be measured are tumor cell killing, T cell activation, relative or absolute T cell number, Fc-mediated effector function (e.g., ADCC, CDC and/or ADCP) or binding to an Fey receptor (FcyR) or FcRn.
  • “Enhanced” may be an enhancement of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more, or a statistically significant enhancement.
  • Fc gamma receptor refers to well-known FcyRI, FcyRIIa, FcyRIIb or FcyRIII. Activating FcyR includes FcyRI, FcyRIIa and FcyRIII.
  • “Fixed dose,” also referred to as “flat dose,” refers to a dose that is administered to a subject without correction for the subject’s specific body weight or body surface area.
  • a fixed dose sometimes referred to as a flat dose, is therefore provided as an absolute amount of the agent (e.g., mg drug), and not as a weight-based amount (e.g. pg/kg or pg drug per kg body weight).
  • a subject weighing 65kg may be administered the same flat dose in milligrams as a subject weighing 85kg.
  • a flat dose may be administered according to a predefined class or category of body weight, but is not modified according to the subject’s specific weight.
  • a “Flat Dose A” may be administered if a patient is greater than a predefined threshold weight (e.g., > 60 kg), whereas a different “Flat Dose B” may be administered if the patent is less than or equal to the pre-defined threshold weight (e.g., ⁇ 60 kg).
  • a predefined threshold weight e.g., > 60 kg
  • a different “Flat Dose B” may be administered if the patent is less than or equal to the pre-defined threshold weight (e.g., ⁇ 60 kg).
  • weight-based refers to administration of a dose amount that is based on the subject’s specific body weight; for example, 3 mg/kg refers to a dose of 3 milligrams of antibody per kilogram of the subject’s body weight. Unless otherwise specified herein, when a dose is described in a unit of “mg/kg” or “pg/kg,” weight-based dosing is being employed.
  • a BCMAxCD3 bispecific antibody such as teclistamab
  • teclistamab is administered on a dosing schedule based on sequential 28-day treatment cycles, for example, Cycle 1 starts on Day 1 of Cycle 1 and ends on Day 28 of Cycle 1, and then Day 1 of Cycle 2 starts the day after Day 28 of Cycle 1 and ends on Day 28 of Cycle 2, and then Day 1 of Cycle 3 starts the day after Day 28 of Cycle 2 and ends on Day 28 of Cycle 3, and so on.
  • Cycle 1 starts on Day 1 of Cycle 1 and ends on Day 28 of Cycle 1
  • Day 1 of Cycle 2 starts the day after Day 28 of Cycle 1 and ends on Day 28 of Cycle 2
  • Day 1 of Cycle 3 starts the day after Day 28 of Cycle 2 and ends on Day 28 of Cycle 3 and so on.
  • Cycles 3-6 (Cycles 3, 4, 5 and 6).
  • a cycle number with a “+” symbol refers to that cycle and all subsequent cycles, e.g., “C3+” refers to from Cycle 3 and all subsequent cycles (i.e., C3, C4, C5, C6, C7, and so on).
  • administration of the BCMAxCD3 bispecific antibody begins in Cycle 1 of a therapeutically effective regimen.
  • administration of the BCMAxCD3 bispecific antibody may start in Cycle 2 or later, i.e., administration of one or more other anti-multiple myeloma agents (but not the BCMAxCD3 bispecific antibody) may start in Cycle 1 and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2 or later.
  • the first treatment cycle that includes administration of the BCMAxCD3 bispecific antibody is Cycle 2 or later.
  • Q4W means once every four weeks (i.e., once every 28 days)
  • Q2W also referred to as “bi-weekly” or “biweekly”
  • QW also referred to as “weekly”
  • Q4W means once weekly.
  • Q4W is also referred to herein as “monthly.”
  • the terms “Q4W” and “monthly” are used interchangeably to refer to once every 4 weeks or once every 28 days (e.g., in sequential 28-day cycles, a first treatment dose occurs on Day 1 of Cycle 1, a second treatment dose occurs on Day 1 of Cycle 2, etc.).
  • Administration of a treatment dose once weekly is also referred to herein as a weekly dosing schedule; for example, a 28-day treatment cycle may have a weekly dosing schedule that comprises four doses one week apart from each other (e.g., on Days 1, 8, 15 and 22), or three doses one week apart from each other (e.g., on Days 8, 15 and 22), or two doses one week apart from each other (e.g., on Days 8 and 15).
  • Administration of a treatment dose once every two weeks (Q2W) is also referred to herein as a bi-weekly dosing schedule.
  • Administration of a treatment dose once every four weeks (Q4W) is also referred to herein as a monthly dosing schedule.
  • Dosing regimens may be described herein in terms of the dose amount and frequency; for example, “Cl: 1500 pg/kg QW” refers to administration of 1500 pg/kg once per week in Cycle 1 of a therapeutically effective regimen, “C3-6: 3000 pg/kg Q2W” refers to administration of 3000 pg/kg once every two weeks from Cycle 3 through Cycle 6, “C3+: 3000 pg/kg Q4W” refers to administration of 3000 pg/kg once every four weeks starting in Cycle 3, etc.
  • CR complete response
  • PR partial response
  • Q2W once every 2 weeks
  • Q4W once every 4 weeks
  • QW once weekly
  • RP2D recommended phase 2 dose
  • SUD step-up dose
  • Human antibody refers to an antibody that is optimized to have minimal immune response when administered to a human subject. Variable regions of human antibody are derived from human immunoglobulin sequences. If human antibody contains a constant region or a portion of the constant region, the constant region is also derived from human immunoglobulin sequences. Human antibody comprises heavy and light chain variable regions that are “derived from” sequences of human origin if the variable regions of the human antibody are obtained from a system that uses human germline immunoglobulin or rearranged immunoglobulin genes. Such exemplary systems are human immunoglobulin gene libraries displayed on phage, and transgenic non-human animals such as mice or rats carrying human immunoglobulin loci.
  • Human antibody typically contains amino acid differences when compared to the immunoglobulins expressed in humans due to differences between the systems used to obtain the human antibody and human immunoglobulin loci, introduction of somatic mutations or intentional introduction of substitutions into the frameworks or CDRs, or both.
  • “human antibody” is at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical in amino acid sequence to an amino acid sequence encoded by human germline immunoglobulin or rearranged immunoglobulin genes.
  • human antibody may contain consensus framework sequences derived from human framework sequence analyses, for example as described in Knappik et al., (2000) J Mol Biol 296:57-86, or synthetic HCDR3 incorporated into human immunoglobulin gene libraries displayed on phage, for example as described in Shi et al., (2010) J Mol Biol 397:385-96, and in Int. Patent Publ. No. W02009/085462.
  • Antibodies in which at least one CDR is derived from a non-human species are not included in the definition of “human antibody”.
  • Humanized antibody refers to an antibody in which at least one CDR is derived from non-human species and at least one framework is derived from human immunoglobulin sequences. Humanized antibody may include substitutions in the frameworks so that the frameworks may not be exact copies of expressed human immunoglobulin or human immunoglobulin germline gene sequences.
  • IMWG diagnostic criteria for multiple myeloma have been met when the participant was diagnosed.
  • IMWG diagnostic criteria are known in the art.
  • Multiple myeloma may be defined as clonal BMPCs >10% or biopsy-proven bony or extramedullary plasmacytoma 3 and at least 1 of the following:
  • Hypercalcemia serum calcium >0.25 mmol/L (>1 mg/dL) higher than the ULN or >2.75 mmol/L (>11 mg/dL)
  • Renal insufficiency creatinine clearance ⁇ 40 mL per min b or serum creatinine >177 pmol/L (>2 mg/dL)
  • Anemia hemoglobin value of >20 g/L below the lower limit of normal, or a hemoglobin value ⁇ 100 g/L
  • Bone lesions 1 or more osteolytic lesions on skeletal radiography, CT, or PET-CT c d
  • isolated refers to a homogenous population of molecules (such as synthetic polynucleotides or a protein such as an antibody) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step.
  • molecules such as synthetic polynucleotides or a protein such as an antibody
  • isolated antibody refers to an antibody that is substantially free of other cellular material and/or chemicals and encompasses antibodies that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity.
  • “Monoclonal antibody” refers to an antibody obtained from a substantially homogenous population of antibody molecules, i.e., the individual antibodies comprising the population are identical except for possible well-known alterations such as removal of C-terminal lysine from the antibody heavy chain or post-translational modifications such as amino acid isomerization or deamidation, methionine oxidation or asparagine or glutamine deamidation.
  • Monoclonal antibodies typically bind one antigenic epitope.
  • a bispecific monoclonal antibody binds two distinct antigenic epitopes.
  • Monoclonal antibodies may have heterogeneous glycosylation within the antibody population.
  • Monoclonal antibody may be monospecific or multispecific such as bispecific, monovalent, bivalent or multivalent.
  • “Mutation” refers to an engineered or naturally occurring alteration in a polypeptide or polynucleotide sequence when compared to a reference sequence. The alteration may be a substitution, insertion or deletion of one or more amino acids or polynucleotides.
  • “Non-fixed combination” refers to separate pharmaceutical compositions of the BCMAxCD3 bispecific antibody and the anti-CD38 antibody administered as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the subject.
  • Multispecific refers to an antibody that specifically binds at least two distinct antigens or at least two distinct epitopes within the same antigen. Multispecific antibody may bind for example two, three, four or five distinct antigens or distinct epitopes within the same antigen.
  • “Pharmaceutical composition” refers to composition that comprises an active ingredient and a pharmaceutically acceptable carrier.
  • “Pharmaceutically acceptable carrier” or “excipient” refers to an ingredient in a pharmaceutical composition, other than the active ingredient, which is nontoxic to a subject.
  • “Philadelphia chromosome” or “Ph” refers to a well-known chromosomal translocation between chromosomes 9 and 22, resulting in the oncogenic BCR-ABL gene fusion with constitutively active tyrosine kinase activity. The translocation results in a portion of the BCR gene from chromosome 22ql 1 becoming fused with a portion of the ABL gene from chromosome 9q34, and is designated as t(9;22)(q34;ql 1) under the International System for Human Cytogenetic Nomenclature (ISCN). Depending on the precise location of the fusion, the molecular weight of the resulting fusion protein can range from 185 to 210 kDa. “Philadelphia chromosome” refers to all BCR-ABL fusion proteins formed due the (9;22)(q34;ql 1) translocation.
  • Recombinant refers to DNA, antibodies and other proteins that are prepared, expressed, created or isolated by recombinant means when segments from different sources are joined to produce recombinant DNA, antibodies or proteins.
  • “Reduce” or “reduced” refers to a reduction in one or more functions of a test molecule when compared to a control molecule or a combination of test molecules when compared to one or more control molecules.
  • Exemplary functions that can be measured are tumor cell killing, T cell activation, relative or absolute T cell number, Fc-mediated effector function (e.g., ADCC, CDC and/or ADCP) or binding to an Fey receptor (FcyR) or FcRn.
  • “Reduced” may be a reduction of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more, or a statistically significant enhancement.
  • rHuPh20 refers to recombinant human hyalurodinase having the amino acid sequence of SEQ ID NO: 5, which is a recombinant hyaluronidase (HYLENEX® recombinant) described in Int'l Pat. Pub. No. W02004/078140.
  • Refractory to a therapy refers to a cancer that is not amendable to surgical intervention and is initially unresponsive to the therapy.
  • Relapsed refers to a cancer that responded to a treatment but then returns.
  • Step-up dose refers to a dose of an active agent that is administered to a subject prior to a treatment dose.
  • a step-up dose is lower than the treatment dose.
  • a “priming” dose strategy may include one or more lower step-up dose(s) followed by higher treatment doses.
  • a “step-up phase” refers to an initial phase of a therapeutically effective regimen in which at least one step-up dose of a therapeutic is administered to the subject.
  • a step-up phase may also include one or more treatment doses, i.e., a step-up phase may include one or more step-up doses followed by one or more treatment doses; for example, a step-up phase may include two step-up doses followed by two treatment doses.
  • the step- up phase is 28 days, i.e., the step-up phase is a 28-day cycle of a therapeutically effective regimen.
  • Subject includes any human or nonhuman animal.
  • Nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Except when noted, the terms “patient” or “subject” are used interchangeably.
  • BCMAxCD3 bispecific antibody refers to a molecule containing two or more binding regions, wherein one of the binding regions specifically binds the cell surface antigen B-cell maturation antigen (BCMA) on a target cell or tissue and wherein a second binding region of the molecule specifically binds a T cell antigen CD3. This dual/multi-target binding ability recruits T cells to the target cell or tissue leading to the eradication of the target cell or tissue.
  • “Therapeutically effective amount” refers to an amount effective, at doses and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount may vary depending on factors such as the disease state, age, sex, and weight of the individual, and the ability of a therapeutic or a combination of therapeutics to elicit a desired response in the individual. Exemplary indicators of an effective therapeutic or combination of therapeutics that include, for example, improved well-being of the patient.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder.
  • Beneficial or desired clinical results include alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if a subject was not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • Treatment dose refers to a dose of the active agent that is administered to a subject to treat a disease.
  • a treatment dose may be administered at a regular dosing interval on a repetitive basis (e.g. weekly, biweekly, monthly).
  • a treatment dose may be preceded by one or more step- up doses.
  • Tumor cell or a “cancer cell” refers to a cancerous, pre-cancerous or transformed cell, either in vivo, ex vivo, or in tissue culture, that has spontaneous or induced phenotypic changes. These changes do not necessarily involve the uptake of new genetic material. Although transformation may arise from infection with a transforming virus and incorporation of new genomic nucleic acid, uptake of exogenous nucleic acid or it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene.
  • Transformation/cancer is exemplified by morphological changes, immortalization of cells, aberrant growth control, foci formation, proliferation, malignancy, modulation of tumor specific marker levels, invasiveness, tumor growth in suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo.
  • suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo.
  • the numbering of amino acid residues in the antibody constant region throughout the specification is according to the EU index as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991), unless otherwise explicitly stated.
  • Antibody constant chain numbering can be found for example at ImMunoGeneTics website, at IMGT Web resources at IMGT Scientific charts.
  • substitutions in the CH3 region are expressed as modified position(s) in the first CH3 domain of the first heavy chain/ modified position(s) in the second CH3 domain of the second heavy chain.
  • F405L/K409R refers to a F405L mutation in the first CH3 region and K09R mutation in the second CH3 region.
  • L351Y_F405A_Y407V/T394W refers to L351Y, F40FA and Y407V mutations in the first CH3 region and T394W mutation in the second CH3 region.
  • D399FHKRQ/K409AGRH refers to mutation in which D399 may be replaced by F, H, K R or Q, and K409 may be replaced by A, G, R or H.
  • the disclosure relates to a method of treating a cancer in a subject in need thereof.
  • the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody, an effective amount of an anti-CD38 antibody, and an effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method may comprise administering any one of a BCMA x CD3 bispecific antibody, an anti-CD38 antibody, and an IMid, or any combination of a BCMA x CD3 bispecific antibody, an anti-CD38 antibody, and an IMid.
  • the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an anti-CD38 antibody to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an IMid to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody and an effective amount of an anti-CD38 antibody to the subject for a time sufficient to treat the cancer.
  • the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an anti-CD38 antibody and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody, an effective amount of an anti-CD38 antibody, and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer.
  • BCMA B-cell maturation antigen
  • BCMA is a cell membrane bound tumor necrosis factor receptor family member involved in differentiation of B-cells to plasma cells. Expression of BCMA is restricted to the B-cell lineage where it is predominantly expressed in the interfollicular region of germinal centers and on differentiated plasma cells and plasmablasts. BCMA is virtually absent on naive and memory B cells.
  • a BCMA x CD3 bispecific antibody targets the CD3 receptor complex on T cells and BCMA on plasma cells. The dual binding site allows the bispecific antibody to draw CD3+ T cells in close proximity to myeloma cells, without regard to T cell receptor specificity or reliance on MHC Class 1 molecules on the surface of antigen presenting cells for activation, leading to cell death of the BCMA-positive cell.
  • any suitable BCMA x CD3 bispecific antibody can be used in any of the embodiments provided herein.
  • Exemplary multispecific and/or bispecific formats include dual targeting molecules including, but not limited to, Dual Targeting (DT)-Ig (GSK/Domantis), Two-in-one Antibody (Genetech) and mAb2 (F-Star), Dual Variable Domain (DVD)-Ig (Abbott), Ts2Ab (Medlmmune/AZ) and BsAb (Zymogenetics), HERCULES (Biogen personal) and TvAb (Roche), ScFv/Fc Fusions (Academic Institution), SCORPION (Emergent BioSolutions/Trubion, Zymogenetics/BMS) and Dual Affinity Retargeting Technology (Fc-DART) (MacroGenics), F(ab)2 (Megarex/AMGEN), Dual-Action or Bis-Fab (Genetech), Dock-and-Lock (DNL) (Immuno
  • the BCMA x CD3 bispecific antibody is an antigen binding fragment.
  • exemplary antigen binding fragments are Fab, F(ab')2, Fd and Fv fragments.
  • the BCMA x CD3 bispecific antibody is chimeric, humanized or human.
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 6, the HCDR2 of SEQ ID NO: 7, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 12, the HCDR2 of SEQ ID NO: 13, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17.
  • the HCDRs and LCDRs of the BCMA x comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 6, the HCDR2 of SEQ ID NO: 7, the HCDR3 of SEQ ID
  • CD3 bispecific antibody are recited in Table 3 below:
  • the CDRs recited in the table above are of the Kabat number system. However, as provided for herein, the CDRs of the present disclosure may be provided by any appropriate numbering system, such as any of the Kabat, Chothia, IMGT or AbM numbering systems. Tables 4-6 below provides exemplary CDRs utilizing the Chothia, AbM, and IMGT numbering systems:
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 46, the HCDR2 of SEQ ID NO: 47, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 48, the HCDR2 of SEQ ID NO: 49, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17.
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 50, the HCDR2 of SEQ ID NO: 51, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 52, the HCDR2 of SEQ ID NO: 53, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17.
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 54, the HCDR2 of SEQ ID NO: 55, the HCDR3 of SEQ ID NO: 56, and a VL having the LCDR1 of SEQ ID NO: 57, a LCDR2 comprising the amino acid sequence DD, and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 58, the HCDR2 of SEQ ID NO: 59, the HCDR3 of SEQ ID NO: 60, and a VL having the LCDR1 of SEQ ID NO: 61, a LCDR2 comprising the amino acid sequence GT and the LCDR3 of SEQ ID NO: 17.
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising the VH of SEQ ID NO: 18 and the VL of SEQ ID NO: 19, and a CD3 binding domain comprising the VH of SEQ ID NO: 20 and the VL of SEQ ID NO: 21.
  • the BCMA x CD3 bispecific antibody that binds BCMA comprises a first heavy chain (HC1) of SEQ ID NO: 22, a first light chain (LC1) of SEQ ID NO: 23, a second heavy chain (HC2) of SEQ ID NO: 24, and a second light chain (LC2) of SEQ ID NO: 25.
  • the BCMA binding arm of the BCMS x CD3 bispecific antibody and the CD3 binging arm of the BCMA x CD3 bispecific antibody comprise the amino acid sequences as provided in Tables 7a and 7b.
  • the BCMA x CD3 bispecific antibody comprises a BCMA binding domain that binds BCMA selected from the group consisting of the BCMA binding domain of ACTR cancer therapy by Seattle Genetics, AFM-26, ALLO-715, anti-BCMA allogenic CAR-T cell therapy by CRISPR Therapeutics, anti-BCMA CAR-T therapy by Sorrento Therapeutics, anti-CD19/BCMA CAR-T cell therapy by Hrain Biotechnology, BCMA CAR-T therapy by Chineo Med (Beijing), BCMA TAC-T cell therapy by Triumvira Immunologies, BCMA-CAR T cell therapy by Shanghai Unicar-Therapy Biomed, BCMA/CD3 antibody by Regeneron, CAR-NK cell therapies by NantKwest, CC-93629, CMD-505, CTX-4419, CY AD- 211, HDP-101, HPN-217, P-BCMA-ALLO1, TNB-383B, bb-2121, AUTO-2, BCMA
  • the BCMA x CD3 bispecific antibody can be, but is not limited to, elranatamab (also named PF-06863135), teneobio (also named TNB-383B), REGN5458, REGN5459, pavurutamab (also named AMG-701), BI 836909, CC-93269, WVT078 or teclistamab (also named JNJ-957 or JNJ-64007957).
  • the BCMA x CD3 bispecific antibody is elranatamab.
  • the BCMA x CD3 bispecific antibody is teneobio.
  • the BCMA x CD3 bispecific antibody is REGN5458.
  • the BCMA x CD3 bispecific antibody is REGN5459. In some embodiments, the BCMA x CD3 bispecific antibody is pavurutamab. In some embodiments, the BCMA x CD3 bispecific antibody is BI 836909. In some embodiments, the BCMA x CD3 bispecific antibody is CC-93269. In some embodiments, the BCMA x CD3 bispecific antibody is WVT078. In some embodiments, the BCMA x CD3 bispecific antibody is teclistamab.
  • teclistamab comprises a first heavy chain (HC1), a first light chain (LC1), a second heavy chain (HC2), and a second light chain (LC2), wherein the HC1 is associated with LC 1 and the HC2 is associated with LC2, wherein HC 1 and LC 1 form a first antigen-binding site that immunospecifically binds to BCMA and wherein HC2 and LC2 form a second antigen-binding site that immunospecifically binds to CD3.
  • HC1 is associated with LC 1
  • HC2 is associated with LC2
  • HC 1 and LC 1 form a first antigen-binding site that immunospecifically binds to BCMA
  • HC2 and LC2 form a second antigen-binding site that immunospecifically binds to CD3.
  • teclistamab comprises a HC1 of SEQ ID NO: 22, a LC1 of SEQ ID NO: 23, a HC2 of SEQ ID NO: 24, and a LC2 of SEQ ID NO: 25.
  • the BCMA arm and the CD3 arm of teclistamab form a functional bispecific antibody through an interaction of their respective Fc domains.
  • the BCMA x CD3 bispecific antibody comprises any one of the BCMA binding domains described in Int. Pat. Publ. No. W02017/031104.
  • the BCMA x CD3 bispecific antibody is an IgGl, an IgG2, an IgG3, or an IgG4 isotype.
  • the BCMA x CD3 bispecific antibody is an IgGl isotype.
  • the BCMA x CD3 bispecific antibody is an IgG2 isotype.
  • the BCMA x CD3 bispecific antibody is an IgG3 isotype.
  • the BCMA x CD3 bispecific antibody is an IgG4 isotype.
  • the BCMA x CD3 bispecific antibody can be of any allotype. It is expected that allotype has no influence on properties of the BCMA x CD3 bispecific antibodies, such as binding or Fc- mediated effector functions. Immunogenicity of therapeutic antibodies is associated with increased risk of infusion reactions and decreased duration of therapeutic response (Baert et al., (2003) N Engl J Med 348:602-08). The extent to which therapeutic antibodies induce an immune response in the host may be determined in part by the allotype of the antibody (Stickler et al., (2011) Genes and Immunity 12:213-21). Antibody allotype is related to amino acid sequence variations at specific locations in the constant region sequences of the antibody. Table 8 shows select IgGl, IgG2, and IgG4 allotypes.
  • the multispecific antibody comprises one or more Fc substitutions that reduces binding of the multispecific antibody to a Fey receptor (FcyR).
  • FcyR Fey receptor
  • substitutions that reduce binding of the multispecific antibody to the FcyR reduces the Fc effector functions such as ADCC, ADCP and/or CDC of the multispecific antibody.
  • the specific substitutions can be made in comparison to the wild-type IgGl of SEQ ID NO: 26 or the wild-type IgG4 of SEQ ID NO: 27.
  • the one or more Fc substitutions is selected from the group consisting of F234A/L235A on IgG4, L234A/L235A on IgGl, V234A/G237A/ P238S/H268A/V309L/A330S/P331S on IgG2, F234A/L235A on IgG4, S228P/F234A/ L235A on IgG4, N297A on all Ig isotypes, V234A/G237A on IgG2, K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl, H268Q/V309L/A330S/P331S on IgG2, S267E/L328F on IgGl, L234F/L235E/D265A on IgGl, L2
  • the one or more Fc substitutions is F234A/L235A on IgG4.
  • the one or more Fc substitutions is L234A/L235A on IgGl. [0113] In some embodiments, the one or more Fc substitutions is V234A/G237A/ P238S/H268A/V309L/A330S/P331S on IgG2.
  • the one or more Fc substitutions is F234A/L235A on IgG4.
  • the one or more Fc substitutions is S228P/F234A/ L235A on IgG4.
  • the one or more Fc substitutions is N297A on all Ig isotypes.
  • the one or more Fc substitutions is V234A/G237A on IgG2.
  • the one or more Fc substitutions is K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl.
  • the one or more Fc substitutions is H268Q/V309L/A330S/ P331S on IgG2.
  • the one or more Fc substitutions is S267E/L328F on IgGl. In some embodiments, the one or more Fc substitutions is L234F/L235E/D265A on IgGl.
  • the one or more Fc substitutions is L234A/L235A/G237A/ P238S/H268A/A330S/P331S on IgGl.
  • the one or more Fc substitutions is S228P/F234A/L235A/ G237A/P238S on IgG4 and S228P/F234A/L235A/G236-deleted/G237A/P238S on IgG4.
  • the bispecific antibody further comprises a S228P substitution.
  • the bispecific antibody comprises one or more asymmetric substitutions in a first CH3 domain or in a second CH3 domain, or in both the first CH3 domain and the second CH3 domain.
  • the one or more asymmetric substitutions is selected from the group consisting of F450L/K409R, wild-type/F409L_R409K, T366Y/F405A, T366W/F405W, F405W/Y407A, T394W/Y407T, T394S/Y407A, T366W/T394S, F405W/T394S and T366W/T366S_L368A_Y407V, L351 Y_F405 A_Y407V/T394W, T366I_K392M_T394W/F405A_Y407V, T366L_K392M_T394W/F405A_Y407V, L351Y_Y407A/T366A_K409F, L351Y_Y407A/T366A_K409F, L351Y_Y407A/T366
  • the one or more asymmetric substitutions is F450L/K409R. In some embodiments, the one or more asymmetric substitutions is wild-type/F409L_R409K. In some embodiments, the one or more asymmetric substitutions is T366Y/F405A. In some embodiments, the one or more asymmetric substitutions is T366W/F405W. In some embodiments, the one or more asymmetric substitutions is F405W/Y407A. In some embodiments, the one or more asymmetric substitutions is T394W/Y407T. In some embodiments, the one or more asymmetric substitutions is T394S/Y407A.
  • the one or more asymmetric substitutions is T366W/T394S. In some embodiments, the one or more asymmetric substitutions is F405W/T394S. In some embodiments, the one or more asymmetric substitutions is T366W/T366S_E368A_Y407V. In some embodiments, the one or more asymmetric substitutions is E351Y_F405A_Y407V/T394W. In some embodiments, the one or more asymmetric substitutions is T366I_K392M_T394W/F405A_Y407V.
  • the one or more asymmetric substitutions is T366E_K392M_T394W/F405A_Y407V. In some embodiments, the one or more asymmetric substitutions is E351Y_Y407A/T366A_K409F. In some embodiments, the one or more asymmetric substitutions is E351Y_Y407A/T366V_K409F. In some embodiments, the one or more asymmetric substitutions is Y407A/T366A_K409F. In some embodiments, the one or more asymmetric substitutions is T366E_K392M_T394W/F405A_Y407V. In some embodiments, the one or more asymmetric substitutions is E351Y_Y407A/T366A_K409F. In some embodiments, the one or more asymmetric substitutions is Y407A/T366A_K409F. In some embodiments, the one or more asymmetric
  • the BCMA x CD3 bispecific antibody is an IgG4 isotype and comprises phenylalanine at position 405 and arginine at position 409 in a first heavy chain (HC1) and leucine at position 405 and lysine at position 409 in a second heavy chain (HC2), wherein residue numbering is according to the EU Index.
  • the BCMA x CD3 bispecific antibody further comprises proline at position 228, alanine at position 234 and alanine at position 235 in both the HC1 and the HC2.
  • BCMA x CD3 bispecific antibody can be any BCMA x CD3 bispecific antibody as provided herein, but it should be understood that the antibody in any of the methods provided for herein can be teclistamab.
  • Anti-CD3 antibodies can be any BCMA x CD3 bispecific antibody as provided herein, but it should be understood that the antibody in any of the methods provided for herein can be teclistamab.
  • CD38 is a multifunctional protein having function in receptor-mediated adhesion and signaling as well as mediating calcium mobilization via its ecto-enzymatic activity, catalyzing formation of cyclic ADP-ribose (cADPR) and AD PR.
  • cADPR cyclic ADP-ribose
  • CD38 mediates cytokine secretion and activation and proliferation of lymphocytes (Funaro et al., J Immunol 145:2390-6, 1990;
  • CD38 via its NAD glycohydrolase activity, also regulates extracellular NAD+ levels, which have been implicated in modulating the regulatory T-cell compartment (Adriouch et al., Microbes infect 14: 1284-92, 2012; Chiarugi et al., Nature Reviews 12:741-52, 2012).
  • CD38 signaling occurs via cross-talk with antigen-receptor complexes on T- and B -cells or other types of receptor complexes, e.g., major histocompatibility complex (MHC) molecules, involving CD38 in several cellular responses, but also in switching and secretion of IgGl.
  • MHC major histocompatibility complex
  • Any suitable anti-CD38 antibody can be used in a method of the application.
  • the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 28, the HCDR2 of SEQ ID NO: 29, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
  • the CDRs recited in above are of the Kabat numbering system. However, as provided for herein, the CDRs of the present disclosure may be provided by any appropriate numbering system, such as any of the Kabat, Chothia, IMGT or AbM numbering systems. Table 9 below provides exemplary CDRs utilizing the Kabat, Chothia, AbM, and IMGT numbering systems:
  • the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 28, the HCDR2 of SEQ ID NO: 29, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
  • the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 62, the HCDR2 of SEQ ID NO: 63, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
  • the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 64, the HCDR2 of SEQ ID NO: 65, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
  • the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 66, the HCDR2 of SEQ ID NO: 67, the HCDR3 of SEQ ID NO: 68, the LCDR1 of SEQ ID NO: 69, an LCDR2 comprising the amino acid sequence of DA, and the LCDR3 of SEQ ID NO: 33.
  • the anti-CD38 antibody comprises the VH of SEQ ID NO: 34 and the VL of SEQ ID NO: 35.
  • the anti-CD38 antibody comprises the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37.
  • anti-CD38 antibodies used in the methods of the invention may be known antibodies, such as mAb003 described in U.S. Pat. No. 7,829,673.
  • the VH and the VL of mAb003 may be expressed as IgGl/K; mAbO24 described in U.S. Pat. No. 7,829,673.
  • the VH and the VL of mAbO24 may be expressed as IgGl/K; MOR-202 (MOR-03087) comprising described in US. Pat. No. 8,088,896.
  • the VH and the VL of MOR-202 may be expressed as IgGl/K; or isatuximab; described in U.S. Pat. No. 8,153,765.
  • the anti-CD38 antibody comprises a) the VH of SEQ ID NO: 38 and the VL of SEQ ID NO: 39; b) the VH of SEQ ID NO: 40 and the VL of SEQ ID NO: 41 ; c) the VH of SEQ ID NO: 42 and the VL of SEQ ID NO: 43; or d) the VH of SEQ ID NO: 44 and the VL of SEQ ID NO: 45.
  • the anti-CD38 antibody is DARZALEX® (daratumumab).
  • daratumumab comprises the VH of SEQ ID NO: 34 and the VL of SEQ ID NO: 35.
  • daratumumab comprises the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37.
  • the anti-CD38 antibody is chimeric, humanized, or human.
  • the anti-CD38 antibody is an IgGl, an IgG2, an IgG3, or an IgG4 isotype.
  • the anti-CD38 antibody is an IgGl isotype.
  • the anti-CD38 antibody is an antigen binding fragment.
  • exemplary antigen binding fragments are Fab, F(ab')2, Fd and Fv fragments.
  • the present application describes various methods utilizing an anti-CD38 antibody. It can be any anti-CD38 antibody as provided herein, but it should be understood that the antibody in any of the methods provided for herein can be daratumumab.
  • Immunomodulatory drugs are known in the art and any appropriate immunomodulatory drug may be used in any of the embodiments provided herein.
  • the immunomodulatory drug is an immunomodulatory imide drug, such as but not limited to thalidomide, lenalidomide, pomalidomide, iberdomide, and apremilast.
  • the immunomodulatory drug is thalidomide.
  • the immunomodulatory drug is lenalidomide.
  • the immunomodulatory drug is pomalidomide.
  • the immunomodulatory drug is iberdomide.
  • the immunomodulatory drug is apremilast.
  • the present application describes various methods utilizing an IMiD. It can be any IMiD as provided herein, but it should be understood that the IMiD in any of the methods provided for herein can be lenalidomide.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any amount administered for a time sufficient to treat the cancer.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 60 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 240 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 355 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 475 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 635 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 720 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 845 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1125 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1500 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1685 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 2250 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 6000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the effective amount of the BCMA x CD3 bispecific antibody is any threshold. In some embodiments, the predetermined weight threshold is 50 kg, 55 kg, 60 kg, 65 kg, 70 kg, greater than 70 kg, or any value in-between.
  • the pre-determined weight threshold is 50 kg, wherein those subjects who are ⁇ 50 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 50 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the pre- determined weight threshold is 55 kg, wherein those subjects who are ⁇ 55 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 55 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 60 kg, wherein those subjects who are ⁇ 60 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 60 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the predetermined weight threshold is 65 kg, wherein those subjects who are ⁇ 65 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 65 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 70 kg, wherein those subjects who are ⁇ 70 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 70 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range inbetween administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • subject is in tier 2 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range inbetween administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is any value including or inbetween the values as provided for herein.
  • the BCMA x CD3 bispecific antibody is any BCMA x CD3 bispecific antibody as provided for herein.
  • the BCMA x CD3 bispecific antibody is teclistamab.
  • teclistamab comprises an amino acid sequence as provided for herein.
  • the therapeutically effective amount of teclistamab is any amount administered for a time sufficient to treat the cancer.
  • the therapeutically effective amount of teclistamab is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 60 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 240 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 300 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 355 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 475 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 635 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 720 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 845 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 1125 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 1500 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 1685 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 2250 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 3000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 6000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is any value including or in-between the values as provided for herein.
  • the therapeutically effective amount of teclistamab is determined based on the weight of the subject. In some embodiments, the therapeutically effective amount of teclistamab is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered a therapeutically effective amount of teclistamab, and wherein if the subject is above the predetermined weight threshold then the subject is in tier 2 and is administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • the therapeutically effective amount of teclistamab for those subjects in tier 1 and the effective amount of teclistamab for those subjects in tier 2 are the same. In some embodiments, the effective amount of teclistamab for those subjects in tier 1 and the effective amount of teclistamab for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the effective amount of teclistamab is any threshold. In some embodiments, the pre-determined weight threshold is 50 kg, 55 kg, 60 kg, 65 kg, 70 kg, greater than 70 kg, or any value in-between.
  • the pre-determined weight threshold is 50 kg, wherein those subjects who are ⁇ 50 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 50 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 55 kg, wherein those subjects who are ⁇ 55 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 55 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 60 kg, wherein those subjects who are ⁇ 60 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 60 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 65 kg, wherein those subjects who are ⁇ 65 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 65 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • the pre-determined weight threshold is 70 kg, wherein those subjects who are ⁇ 70 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 70 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
  • subject is in tier 1 and the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 1 and the therapeutically effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is any value including or in-between the values as provided for herein.
  • subject is in tier 2 and the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
  • the subject is in tier 2 and the effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is any value including or in-between the values as provided for herein.
  • the present application describes various methods utilizing a BCMA x CD3 bispecific antibody. It can be any BCMA x CD3 bispecific antibody as provided herein, but it should be understood that the antibody in any of the methods can be teclistamab.
  • the therapeutically effective amount of the anti-CD38 antibody is any amount administered for a time sufficient to treat the cancer.
  • the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg, or any value or range in-between administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the anti-CD38 antibody is 1200 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the anti-CD38 antibody is 1300 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the anti-CD38 antibody is 1400 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1500 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1600 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1700 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the anti-CD38 antibody is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1900 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2000 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2100 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the anti-CD38 antibody is 2200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2400 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the anti-CD38 antibody is FDA-approved for clinical use. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the anti-CD38 antibody can be any anti-CD38 antibody as provided for herein.
  • the anti-CD38 antibody is daratumumab.
  • daratumumab comprises the amino acid sequences as provided for herein.
  • the therapeutically effective amount of daratumumab is any amount administered for a time sufficient to treat the cancer.
  • the therapeutically effective amount of daratumumab is from about 1200 mg to about 2400 mg, or any value or range in-between administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of daratumumab is 1200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1400 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1500 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of daratumumab is 1600 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1700 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1900 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of daratumumab is 2000 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2100 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2400 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of daratumumab is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of daratumumab is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks.
  • the present application describes various methods utilizing an anti-CD38 antibody. It can be any anti-CD38 antibody as provided herein, but it should be understood that the antibody in any of the methods can be daratumumab. IMiD dosing
  • the therapeutically effective amount of the IMiD is any amount administered for a time sufficient to treat the cancer.
  • the effective amount of the IMiD is from about 15 mg to about 50 mg, or any value or range in-between administered once a day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of the IMiD is 25 mg administered daily.
  • the IMiD is FDA approved for clinical use.
  • the therapeutically effective amount of the IMiD is based on the standard doses used in clinical practices for the treatment of cancer.
  • the therapeutically effective amount of the IMiD is 25 mg.
  • the IMiD is any IMiD as provided for herein.
  • the IMiD is lenalidomide.
  • the therapeutically effective amount of lenalidomide is any amount administered for a time sufficient to treat the cancer.
  • the effective amount of lenalidomide is from about 15 mg to about 50 mg, or any value or range in-between administered once a day, once a week, biweekly, triweekly, or once every four weeks.
  • the therapeutically effective amount of lenalidomide is 25 mg administered daily.
  • the therapeutically effective amount of lenalidomide is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of lenalidomide is 25 mg.
  • the BCMA x CD3 bispecific antibody, anti-CD38 antibody, and IMiD of the methods disclosed herein may be administered to the subject via any appropriate method of administration.
  • Exemplary methods of administration include, but are not limited to transarterial, subcutaneous, intradermal, intratumoral, intraanodal, intramedullary, intramuscular, intravenous, intraperitoneal, intranasal, or intraosseous.
  • the BCMA x CD3 bispecific antibody is administered intravenously or subcutaneously.
  • the BCMA x CD3 bispecific antibody is administered intravenously (i.v.).
  • the BCMA x CD3 bispecific antibody is administered subcutaneously (s.c.).
  • the anti-CD38 bispecific antibody is administered intravenously or subcutaneously. In some embodiments, the anti-CD38 bispecific antibody is administered intravenously (i.v.). In some embodiments, the anti-CD38 bispecific antibody is administered subcutaneously (s.c.). In some embodiments, the IMiD bispecific antibody is administered intravenously or orally. In some embodiments, the IMiD bispecific antibody is administered intravenously (i.v.). In some embodiments, the IMiD bispecific antibody is administered orally.
  • the method comprises intravenously administering an therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises intravenously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises intravenously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) (oral) to the subject for a time sufficient to treat the cancer.
  • IMD immunomodulatory drug
  • the components of the methods provided for herein may be provided at any dose or frequency required to effectively treat the cancer.
  • the therapeutically effective amount of the components of the methods provided for herein may be provided at any frequency required to effectively treat the cancer.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered weekly. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered biweekly. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once every three weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once a month.
  • the frequency of administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be altered as necessary to effectively treat the cancer.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
  • the therapeutically effective amount of the anti-CD38 antibody is administered daily. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered weekly. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered biweekly. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered once every three weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered once every four weeks. In some embodiments, the therapeutically effective amount of the anti- CD38 antibody is administered once a month. The frequency of administration of the therapeutically effective amount of the anti-CD38 antibody may be altered as necessary to effectively treat the cancer.
  • the therapeutically effective amount of the anti-CD38 antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the anti-CD38 antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
  • the therapeutically effective amount of the IMiD is administered daily. In some embodiments, the therapeutically effective amount of the IMiD is administered weekly. In some embodiments, the therapeutically effective amount of the IMiD is administered biweekly. In some embodiments, the therapeutically effective amount of the IMiD is administered once every three weeks. In some embodiments, the therapeutically effective amount of the IMiD is administered once every four weeks. In some embodiments, the therapeutically effective amount of the IMiD is administered once a month. The frequency of administration of the therapeutically effective amount of the IMiD may be altered as necessary to effectively treat the cancer.
  • the therapeutically effective amount of the IMiD may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the IMiD may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
  • the therapeutically effective amount and frequency of a component of the method is held constant for a recurring period of time, or a cycle.
  • the duration of a treatment cycle may be for any period of time.
  • a treatment cycle may last for 7, 8, 9, 10, 11, 12, 13, 14, 21, 28, 56, 112, 224, 365, more than 365 days, or any number of days in-between.
  • the treatment cycle lasts for 28 days. It is to be understood that the length of time of a treatment cycle is not meant to limit the duration of the treatment in any way. The treatment may continue for any number of treatment cycles necessary to treat the cancer.
  • the components of the methods provided for herein may be administered to the subject for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more than 20 treatment cycles, or the components of the methods provided for herein are administered to the subject until the cancer is treated.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for one treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for two treatment cycles.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for three treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for four treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for five treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for six treatment cycles.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for seven treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for more than seven treatment cycles. Further, it is to be understood, as provided for herein, that the therapeutically effective amount or frequency of the components of the methods provided for herein may be altered at the conclusion of each treatment cycle. Accordingly, in some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for one treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for two treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for three treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti- CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for four treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for five treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for six treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for seven treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti- CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for more than seven treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered weekly for a first and second treatment cycles, biweekly for a third, fourth, fifth, and sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles.
  • the therapeutically effective amount of the anti-CD38 antibody is administered weekly for a first and second treatment cycles, biweekly for a third, fourth, fifth, and sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles.
  • the therapeutically effective amount of the IMiD is administered daily for days 1-21 of a 28-day treatment cycle.
  • the therapeutically effective amount of the IMiD is not administered in treatment cycle one.
  • the therapeutically effective amount of the IMiD is administered starting at treatment cycle two.
  • the duration of the treatment of the method may continue for any amount of time necessary to treat the cancer.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, 48, 52, 104, 156, 208, 260 or more weeks, or any number of weeks in-between.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 260 weeks.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or more months, or any number of months in-between.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 60 months.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years, or any number of years inbetween.
  • the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 10 years.
  • components of the methods provided for herein may be provided as a loading dose, a treatment dose, or a combination thereof. In some embodiments, components of the methods provided for herein are provided at a treatment dose. In some embodiments, components of the methods provided for herein are provided at a loading dose followed by a treatment dose. In some embodiments, the loading dose comprises one or more step-up doses provided over a period of time. In some embodiments, one step-up dose is provided. In some embodiments, two step-up doses are provided. In some embodiments, three step-up doses are provided. In some embodiments, more than three step-up doses are provided. In some embodiments, any number of step-up doses may be provided as necessary in order to provide the subject the effective amount of the components necessary to treat the cancer.
  • the BCMA x CD3 bispecific antibody may be administered at a loading dose followed by a treatment dose.
  • the loading dose comprises one or more step-up doses as provided for herein.
  • the BCMA x CD3 bispecific antibody is administered as one step-up dose followed by a treatment dose.
  • the BCMA x CD3 bispecific antibody is administered as two step-up doses followed by a treatment dose.
  • the BCMA x CD3 bispecific antibody is administered as three step-up doses followed by a treatment dose.
  • the BCMA x CD3 bispecific antibody is administered as more than three step-up doses followed by a treatment dose.
  • a first step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 300 Pg/kg-
  • the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the first step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an first step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an first step-up dose of the BCMA x CD3 bispecific antibody, wherein the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the first step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
  • subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • second step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 300 pg/kg.
  • the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the second step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an second step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an second step-up dose of the BCMA x CD3 bispecific antibody, wherein the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the second step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
  • subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 300 pg/kg.
  • the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the third or greater step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is determined based on a predetermined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an third or greater step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an third or greater step-up dose of the BCMA x CD3 bispecific antibody, wherein the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
  • the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the third or greater step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
  • subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range inbetween. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg.
  • the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range inbetween. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg.
  • the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose A and comprise a dose from about 60 pg/kg to about 150 pg/kg, or any value or range in-between.
  • the Step-up dose A of the BCMA x CD3 bispecific antibody is 60 pg/kg.
  • the Step-up dose A of the BCMA x CD3 bispecific antibody is 70 pg/kg.
  • the Step-up dose A of the BCMA x CD3 bispecific antibody is 80 pg/kg.
  • the Step-up dose A of the BCMA x CD3 bispecific antibody is 90 pg/kg.
  • the Step-up dose A of the BCMA x CD3 bispecific antibody is 100 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 125 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 150 pg/kg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose B and comprise a dose from about 150 pg/kg to about 250 or any value or range in-between.
  • the Step-up dose B of the BCMA x CD3 bispecific antibody is 150 pg/kg.
  • the Step-up dose B of the BCMA x CD3 bispecific antibody is 160 pg/kg.
  • the Step-up dose B of the BCMA x CD3 bispecific antibody is 170 pg/kg.
  • the Step-up dose B of the BCMA x CD3 bispecific antibody is 180 pg/kg.
  • the Step-up dose B of the BCMA x CD3 bispecific antibody is 190 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 200 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 210 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 220 pg/kg. In some embodiments, the Step- up dose B of the BCMA x CD3 bispecific antibody is 230 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 240pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 250pg/kg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose C and comprise a dose from about 5 pg/kg to about 60 pg/kg, or any value or range in-between.
  • the Step-up dose C of the BCMA x CD3 bispecific antibody IS 5 pg/kg.
  • the Step-up dose C of the BCMA x CD3 bispecific antibody IS 10 pg/kg.
  • the Step-up dose C of the BCMA x CD3 bispecific antibody IS 15 pg/kg.
  • the Step-up dose C of the BCMA x CD3 bispecific antibody IS 20 pg/kg.
  • the Step-up dose C of the BCMA x CD3 bispecific antibody IS 25 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 30 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 40 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody is 50 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody is 60 pg/kg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose D and comprise a dose from about 250 pg/kg to about 600 pg/kg, or any value or range in-between.
  • the Step-up dose D of the BCMA x CD3 bispecific antibody is 250 pg/kg.
  • the Step-up dose D of the BCMA x CD3 bispecific antibody is 300 pg/kg.
  • the Step-up dose D of the BCMA x CD3 bispecific antibody is 350 pg/kg.
  • the Step-up dose D of the BCMA x CD3 bispecific antibody is 400 pg/kg.
  • the Step-up dose D of the BCMA x CD3 bispecific antibody is 450 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 500 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 550 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 600 pg/kg. [0201] In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody of the methods provided for herein may comprise one or more of any dose or range selected from Step- up Dose A, B, C, D, or any combination thereof. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up doses as provided for in Table 10 below:
  • a step-up doses of the BCMA x CD3 bispecific antibody may be designated as Step-up dose E and comprise a dose from about 3 mg to about 12 mg, or any value or range in-between.
  • the Step-up dose E of the BCMA x CD3 bispecific antibody is 3 mg.
  • the Step-up dose E of the BCMA x CD3 bispecific antibody is 4 mg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose F and comprise a dose from about 12 mg to about 20 mg, or any value or range in-between.
  • the Step-up dose F of the BCMA x CD3 bispecific antibody is 12 mg.
  • the Step-up dose F of the BCMA x CD3 bispecific antibody is 15 mg.
  • the Step-up dose F of the BCMA x CD3 bispecific antibody is 20 mg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose G and comprise a dose from about 20 mg to about 30 mg, or any value or range in-between.
  • the Step-up dose G of the BCMA x CD3 bispecific antibody is 20 mg.
  • the Step-up dose G of the BCMA x CD3 bispecific antibody is 24 mg.
  • the Step-up dose G of the BCMA x CD3 bispecific antibody is 25 mg.
  • the Step-up dose G of the BCMA x CD3 bispecific antibody is 30 mg.
  • a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose H and comprise a dose from about 30 mg to about 50 mg, or any value or range in-between.
  • the Step-up dose H of the BCMA x CD3 bispecific antibody is 30 mg.
  • the Step-up dose H of the BCMA x CD3 bispecific antibody is 40 mg.
  • the Step-up dose H of the BCMA x CD3 bispecific antibody is 50 mg.
  • the loading dose of the BCMA x CD3 bispecific antibody of the methods provided for herein may comprise one or more of any dose or range selected from Step- up Dose E, F, G, or H, or any combination thereof.
  • the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up doses as provided for in Table 11 below:
  • the current step-up dose and subsequent step-up does(s) may comprise the same or different doses and may comprises the same or different frequencies.
  • the current step-up dose and subsequent step-up dose(s) comprise the same dose and same frequency.
  • the current step-up dose and subsequent step-up dose(s) comprise the same dose and different frequencies.
  • the current step-up dose and subsequent step-up dose(s) comprise different doses and the same frequency.
  • the current step-up dose and subsequent step-up dose(s) comprise different doses and different frequencies.
  • the loading dose of the BCMA x CD3 bispecific antibody is administered at a frequency as provided for herein. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered every other day. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered weekly.
  • the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up dose(s). Accordingly, in some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered at a frequency as provided for herein. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered daily. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered every other day. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered weekly.
  • the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 10.
  • the one or more step-up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered daily.
  • the one or more step- up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered every other day.
  • the one or more step-up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered weekly.
  • each step-up dose A, B, C, or D may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein.
  • a first step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group A, B, C, or D as the first step-up dose and wherein the first and second time periods may be the same or different.
  • a third step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a third time period, wherein the first, second, and third step-up doses are not of the same group A, B, C, or D and wherein the first, second, and third time periods may be the same or different.
  • a fourth step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a fourth time period, wherein the first, second, third, and fourth step-up doses are all of a unique group A, B, C, or D and wherein the first, second, third, and fourth time periods may be the same or different.
  • the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 11.
  • the one or more step-up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered daily.
  • the one or more step- up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered every other day.
  • the one or more step-up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered weekly.
  • each step-up dose E, F, G, or H may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein. Accordingly, in some embodiments, a first step-up dose selected from E,
  • F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group E, F,
  • a third step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly month for a third time period, wherein the first, second, and third step-up doses are not of the same group E, F, G, or H and wherein the first, second, and third time periods may be the same or different.
  • a fourth step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a fourth time period, wherein the first, second, third, and fourth step-up doses are not of the same group E, F, G, or H and wherein the first, second, third, and fourth time periods may be the same or different.
  • treatment dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 300 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 355 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 475 pg/kg.
  • the treatment dose of the BCMA x CD3 bispecific antibody is 635 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 720 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 845 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1125 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1500 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1685 pg/kg.
  • the treatment dose of the BCMA x CD3 bispecific antibody is 2250 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 3000 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 6000 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg.
  • the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • the treatment dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an treatment dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an treatment dose of the BCMA x CD3 bispecific antibody, wherein the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same.
  • the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different.
  • the predetermined weight threshold to determine the treatment dose of the BCMA x CD3 bispecific antibody is as provided for herein.
  • subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between.
  • the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg.
  • the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg.
  • the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg.
  • the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between.
  • the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg.
  • the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg.
  • the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg.
  • the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg.
  • the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
  • the treatment dose of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered weekly. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered biweekly. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once every three weeks. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once every four weeks. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once a month. The frequency of administration of the treatment dose of the BCMA x CD3 bispecific antibody may be altered as necessary to treat the cancer.
  • the treatment dose of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the treatment dose of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
  • the method of treating cancer comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen, a cotreatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer.
  • the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti- CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen.
  • the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a cotreatment regimen.
  • the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen and a co-treatment regimen.
  • any of the methods of the present disclosure may further comprise the administration of a pre-treatment regimen, a co-treatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer.
  • a method as provided for herein further comprises administering a pre-treatment regimen, a co-treatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer.
  • a method as provided for herein further comprises administering a pre-treatment regimen for a time sufficient to treat the cancer.
  • a method as provided for herein further comprises administering a co-treatment regimen for a time sufficient to treat the cancer.
  • a method as provided for herein further comprises administering a pre-treatment regimen and a co-treatment regimen to the subject for a time sufficient to treat the cancer.
  • pre-treatment regimen can refer to a regimen of pharmaceutical compounds, compositions, or formulas that are administered prior to the start of the administration of a therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD).
  • a therapeutically effective amount of the BCMA x CD3 bispecific antibody a therapeutically effective amount of the anti-CD38 antibody
  • IMD immunomodulatory drug
  • Pre-treatment regimen can also refer to refer to a regimen of pharmaceutical compounds, compositions, or formulas that are administered during the course of administration of a therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD), but wherein the pre-treatment regimen is administered prior to administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti- CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD) should dosing occur on the same day.
  • IMD immunomodulatory drug
  • pretreatment regimen dose not exclude the components of the pre-treatment regimen from be administered during the course of administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD).
  • IMD immunomodulatory drug
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, a therapeutically effective amount of an antipyretic, or any combination thereof.
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid.
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine.
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of an antipyretic.
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, and a therapeutically effective amount of an antihistamine.
  • the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid and a therapeutically effective amount of an antipyretic. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine and a therapeutically effective amount of an antipyretic. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, and a therapeutically effective amount of an antipyretic.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, a therapeutically effective amount of an antipyretic, or any combination thereof.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of an antipyretic.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, and a therapeutically effective amount of an antihistamine.
  • the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid and a therapeutically effective amount of an antipyretic. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine and a therapeutically effective amount of an antipyretic. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, and a therapeutically effective amount of an antipyretic.
  • the therapeutically effective amount of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer.
  • the therapeutically effective amount of the glucocorticoid is from about 8 mg to about 50 mg or any value or range in-between.
  • the therapeutically effective amount of the glucocorticoid is 8 mg.
  • the therapeutically effective amount of the glucocorticoid is 10 mg.
  • the therapeutically effective amount of the glucocorticoid is 12 mg.
  • the therapeutically effective amount of the glucocorticoid is 14 mg.
  • the therapeutically effective amount of the antihistamine is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer.
  • the therapeutically effective amount of the antihistamine is from about 25 mg to about 50 mg, or any value or range in-between, In some embodiments, the therapeutically effective amount of the antihistamine is 25 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 30 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 35 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 40 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 45 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 50 mg.
  • the therapeutically effective amount of the antipyretic is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer.
  • the therapeutically effective amount of the antipyretic is from about 500 mg to about 1000 mg, or any value or range in-between.
  • the therapeutically effective amount of the antipyretic is 500 mg.
  • the therapeutically effective amount of the antipyretic is 550 mg.
  • the therapeutically effective amount of the antipyretic is 600 mg.
  • the therapeutically effective amount of the antipyretic is 650 mg.
  • the therapeutically effective amount of the antipyretic is 700 mg. some embodiments, the therapeutically effective amount of the antipyretic is 750 mg.
  • the therapeutically effective amount of the antipyretic is 800 mg. some embodiments, the therapeutically effective amount of the antipyretic is 850 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 900 mg. some embodiments, the therapeutically effective amount of the antipyretic is 950 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 1000 mg.
  • the glucocorticoid, antihistamine, and antipyretic of the methods disclosed herein may be administered to the subject via any appropriate method of administration.
  • Exemplary methods of administration include, but are not limited to transarterial, subcutaneous, intradermal, intratumoral, intraanodal, intramedullary, intramuscular, intravenous, intraperitoneal, intranasal, or intraosseous.
  • the glucocorticoid is administered orally or intravenously (i.v.).
  • the glucocorticoid is administered orally.
  • the glucocorticoid is administered intravenously (i.v.).
  • the antihistamine is administered orally or intravenously (i.v.). In some embodiments, the antihistamine is administered orally. In some embodiments, the antihistamine is administered intravenously (i.v.). In some embodiments, the antipyretic is administered orally or intravenously (i.v.). In some embodiments, the antipyretic is administered orally. In some embodiments, the antipyretic is administered intravenously (i.v.).
  • the components of the pre-treatment and co-treatment regimens of the methods provided for herein may be provided at any dose or frequency.
  • the therapeutically effective amount of the pre-treatment and co-treatment regimens may be provided at any frequency.
  • the glucocorticoid is administered daily. In some embodiments, effective amount of the glucocorticoid is administered every other day. In some embodiments, effective amount of the glucocorticoid is administered weekly. In some embodiments, effective amount of the glucocorticoid is administered biweekly. In some embodiments, effective amount of the glucocorticoid is administered once every three weeks. In some embodiments, effective amount of the glucocorticoid is administered once every four weeks. In some embodiments, effective amount of the glucocorticoid is administered once a month. The frequency of administration of the effective amount of the glucocorticoid may be altered as necessary to achieve a desired effect.
  • the effective amount of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time.
  • the antihistamine is administered daily. In some embodiments, effective amount of the antihistamine is administered every other day. In some embodiments, effective amount of the antihistamine is administered weekly. In some embodiments, effective amount of the antihistamine is administered biweekly. In some embodiments, effective amount of the antihistamine is administered once every three weeks. In some embodiments, effective amount of the antihistamine is administered once every four weeks. In some embodiments, effective amount of the antihistamine is administered once a month. The frequency of administration of the effective amount of the antihistamine may be altered as necessary to achieve a desired effect.
  • the effective amount of the antihistamine may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the antihistamine may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time.
  • the antipyretic is administered daily. In some embodiments, effective amount of the antipyretic is administered every other day. In some embodiments, effective amount of the antipyretic is administered weekly. In some embodiments, effective amount of the antipyretic is administered biweekly. In some embodiments, effective amount of the antipyretic is administered once every three weeks. In some embodiments, effective amount of the antipyretic is administered once every four weeks. In some embodiments, effective amount of the antipyretic is administered once a month. The frequency of administration of the effective amount of the antipyretic may be altered as necessary to achieve a desired effect.
  • the effective amount of the antipyretic may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the antipyretic may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time.
  • the therapeutically effective amount and frequency of a component of the method is held constant for a recurring period of time, or a cycle.
  • the duration of a treatment cycle may be for any period of time.
  • a treatment cycle may last for 7, 8, 9, 10, 11, 12, 13, 14, 21, 28, 56, 112, 224, 365, more than 365 days, or any number of days in-between. In some embodiments, the treatment cycle lasts for 28 days. It is to be understood that the length of time of a treatment cycle is not meant to limit the duration of the treatment in any way.
  • the treatment may continue for any number of treatment cycles necessary to treat the cancer.
  • the components of the methods provided for herein may be administered to the subject for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more than 20 treatment cycles, or the components of the methods provided for herein are administered to the subject until the cancer is treated.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for two treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for three treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for four treatment cycles.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for five treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for six treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for seven treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for more than seven treatment cycles.
  • any of the components herein may or may not be administered in any of the treatment cycles described herein.
  • the therapeutically effective amount of the glucocorticoid may be administered for a first number of treatment cycles as provided for herein
  • the therapeutically effective amount of the antihistamine may be administered for a second number of treatment cycles
  • the therapeutically effective amount of the antipyretic may be administered for a third number of treatment cycles, wherein the first, second and third number of treatment cycles may be the same or different.
  • the effective amount or frequency of the components of the methods provided for herein may be altered at the conclusion of each treatment cycle. Accordingly, in some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for two treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for three treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for four treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for five treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for six treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
  • the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for seven treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for more than seven treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
  • the therapeutically effective amount of the glucocorticoid is administered on days 1, 2, 4, and 8 of a first treatment cycle, and then administered weekly for a second, third, and fourth treatment cycle.
  • the therapeutically effective amount of the antihistamine is administered weekly for a first and second treatment cycles, and then administered biweekly for a third, fourth, fifth, and sixth treatment cycle.
  • the therapeutically effective amount of the antipyretic is administered weekly for a first and second treatment cycles, and then administered biweekly for a third, fourth, fifth, and sixth treatment cycle.
  • the therapeutically effective amount of the glucocorticoid is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered for a first, second, third, and fourth treatment cycles. In some embodiments, the therapeutically effective amount of the glucocorticoid is further administered after an administration relation reaction to the anti-CD38 antibody. In some embodiments, the therapeutically effective amount of the glucocorticoid is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody. In some embodiments, the therapeutically effective amount of the antihistamine is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered.
  • the therapeutically effective amount of the antihistamine is further administered after an administration relation reaction to the anti- CD38 antibody. In some embodiments, the therapeutically effective amount of the antihistamine is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody. In some embodiments, the therapeutically effective amount of the antipyretic is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered. In some embodiments, the therapeutically effective amount of the antipyretic is further administered after an administration relation reaction to the anti-CD38 antibody. In some embodiments, the therapeutically effective amount of the antipyretic is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody.
  • the duration of the treatment of the method may continue for any amount of time necessary to treat the cancer.
  • the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, 48, 52, 104, 156, 208, 260 or more weeks, or any number of weeks inbetween.
  • the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 260 weeks.
  • the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or more months, or any number of months in-between. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 60 months. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years, or any number of years in-between. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 10 years.
  • components of the method provided for herein may be provided as a loading dose, a treatment dose, or a combination thereof.
  • components of the methods provided for herein are provided at a treatment dose.
  • components of the methods provided for herein are provided at a loading dose followed by a treatment dose.
  • the loading dose comprises one or more step-up doses provided over a period of time.
  • one step-up dose is provided.
  • two step-up doses are provided.
  • three step-up doses are provided.
  • four step-up doses are provided. In some embodiments, more than four step-up doses are provided.
  • the glucocorticoid may be administered at a loading dose followed by a treatment dose.
  • the loading dose comprises one or more step-up doses as provided for herein.
  • the glucocorticoid is administered as one step-up dose followed by a treatment dose.
  • the glucocorticoid is administered as two step-up doses followed by a treatment dose.
  • the glucocorticoid is administered as three step-up doses followed by a treatment dose.
  • the glucocorticoid is administered as four step-up doses followed by a treatment dose.
  • the first step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the first step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the first step-up dose of the glucocorticoid is 8 mg. In some embodiments, the first step-up dose of the glucocorticoid is 10 mg. In some embodiments, the first step-up dose of the glucocorticoid is 12 mg. In some embodiments, the first step-up dose of the glucocorticoid is 14 mg.
  • the first step-up dose of the glucocorticoid is 16 mg. In some embodiments, the first step-up dose of the glucocorticoid is 18 mg. In some embodiments, the first step-up dose of the glucocorticoid is 20 mg. In some embodiments, the first step-up dose of the glucocorticoid is 25 mg. In some embodiments, the first step-up dose of the glucocorticoid is 30 mg. In some embodiments, the first step-up dose of the glucocorticoid is 35 mg. In some embodiments, the first step-up dose of the glucocorticoid is 40 mg. In some embodiments, the first step-up dose of the glucocorticoid is 45 mg. In some embodiments, the first step-up dose of the glucocorticoid is 50 mg.
  • the second step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer.
  • the second step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between.
  • the second step-up dose of the glucocorticoid is 8 mg.
  • the second step-up dose of the glucocorticoid is 10 mg.
  • the second step-up dose of the glucocorticoid is 12 mg.
  • the second step-up dose of the glucocorticoid is 14 mg.
  • the second step-up dose of the glucocorticoid is 16 mg. In some embodiments, the second step-up dose of the glucocorticoid is 18 mg. In some embodiments, the second step-up dose of the glucocorticoid is 20 mg. In some embodiments, the second step-up dose of the glucocorticoid is
  • the second step-up dose of the glucocorticoid is 30 mg. In some embodiments, the second step-up dose of the glucocorticoid is 35 mg. In some embodiments, the second step-up dose of the glucocorticoid is 40 mg. In some embodiments, the second step-up dose of the glucocorticoid is 45 mg. In some embodiments, the second step-up dose of the glucocorticoid is 50 mg.
  • the third, fourth, or greater step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer.
  • the third, fourth or greater step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between.
  • the third, fourth, or greater step-up dose of the glucocorticoid is 8 r ig.
  • the third, fourth, or greater step-up dose of the glucocorticoid is 10 mg.
  • the third, fourth, or greater step-up dose of the glucocorticoid is 12 mg.
  • the third, fourth, or greater step-up dose of the glucocorticoid is 14 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 16 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 18 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 20 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 25 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 30 mg.
  • the third, fourth, or greater step-up dose of the glucocorticoid is 35 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 40 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 45 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 50 mg.
  • a step-up dose of the glucocorticoid may be designated as Step-up dose J and comprise a dose from about, about 8 mg to about 12 mg, or any value or range inbetween.
  • the Step-up dose J of the glucocorticoid is 8 mg.
  • the Step-up dose J of the glucocorticoid is 9 mg.
  • the Step- up dose J of the glucocorticoid is 10 mg.
  • the Step-up dose J of the glucocorticoid is 11 mg.
  • the Step-up dose J of the glucocorticoid is 12 mg.
  • a step-up dose of the glucocorticoid may be designated as Step-up dose K and comprise a dose from about 12 mg to about 20 mg, or any value or range in-between.
  • the Step-up dose K of the glucocorticoid is 12 mg.
  • the Step-up dose K of the glucocorticoid is 14 mg.
  • the Step-up dose K of the glucocorticoid is 16 mg.
  • the Step-up dose K of the glucocorticoid is 18 mg.
  • the Step-up dose K of the glucocorticoid is 20 mg.
  • a step-up dose of the glucocorticoid may be designated as Step-up dose L and comprise a dose from about 20 mg to about 50 mg, or any value or range in-between.
  • the Step-up dose L of the glucocorticoid is 20 mg.
  • the Step-up dose L of the glucocorticoid is 25 mg.
  • the Step-up dose L of the glucocorticoid is 30 mg.
  • the Step-up dose L of the glucocorticoid is 35 mg.
  • the Step-up dose L of the glucocorticoid is 40 mg.
  • the Step-up dose L of the glucocorticoid is 45 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 50 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 60 mg. In some
  • the loading dose of the glucocorticoid of the methods provided for herein may comprise one or more of any dose or range selected from Step-up Dose J, K, L, or any combination thereof.
  • the loading dose of the glucocorticoid may comprise one or more step-up doses as provided for in Table 12 below:
  • the current step-up dose and subsequent step-up does(s) may comprise the same or different doses and may comprises the same or different frequencies.
  • the current step-up dose and subsequent step-up dose(s) comprise the same dose and same frequency.
  • the current step-up dose and subsequent step-up dose(s) comprise the same dose and different frequencies.
  • the current step-up dose and subsequent step-up dose(s) comprise different doses and the same frequency.
  • the current step-up dose and subsequent step-up dose(s) comprise different doses and different frequencies.
  • the loading dose of the glucocorticoid is administered at a frequency as provided for herein. In some embodiments, the loading dose of the glucocorticoid is administered daily. In some embodiments, the loading dose of the glucocorticoid is administered every other day. In some embodiments, the loading dose of the glucocorticoid is administered weekly.
  • the loading dose of the glucocorticoid may comprise one or more step-up dose(s). Accordingly, in some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered at a frequency as provided for herein. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered daily. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered every other day. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered weekly.
  • the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 12.
  • the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered daily.
  • the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered every other day.
  • the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered weekly.
  • each step-up dose J, K, or L may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein.
  • a first step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group J, K, or L as the first step-up dose and wherein the first and second time periods may be the same or different.
  • a third step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a third time period, wherein the first, second, and third step-up doses are not of the same group J, K, or L and wherein the first, second, and third time periods may be the same or different.
  • the treatment dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the treatment dose of the glucocorticoid is 8 mg. In some embodiments, the treatment dose of the glucocorticoid is 10 mg. In some embodiments, the treatment dose of the glucocorticoid is 12 mg. In some embodiments, the treatment dose of the glucocorticoid is 14 mg. In some embodiments, the treatment dose of the glucocorticoid is 16 mg. In some embodiments, the treatment dose of the glucocorticoid is 18 mg.
  • the treatment dose of the glucocorticoid is 20 mg. In some embodiments, the treatment dose of the glucocorticoid is 25 mg. In some embodiments, the treatment dose of the glucocorticoid is 30 mg. In some embodiments, the treatment dose of the glucocorticoid is 35 mg. In some embodiments, the treatment dose of the glucocorticoid is 40 mg. In some embodiments, the treatment dose of the glucocorticoid is 45 mg. In some embodiments, the treatment dose of the glucocorticoid is 50 mg.
  • the treatment dose of the glucocorticoid is administered daily. In some embodiments, the treatment dose of the glucocorticoid is administered weekly. In some embodiments, the treatment dose of the glucocorticoid is administered biweekly. In some embodiments, the treatment dose of the glucocorticoid is administered once every three weeks. In some embodiments, the treatment dose of the glucocorticoid is administered once every four weeks. In some embodiments, the treatment dose of the glucocorticoid is administered once a month. The frequency of administration of the treatment dose of the glucocorticoid may be altered as necessary to achieve the desired effect.
  • the treatment dose of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the treatment dose of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
  • the components of the pre-treatment regimen or the co-treatment regimen are administered prior to the start of administering the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD).
  • the components of the pre-treatment regimen or the co-treatment regimen are administered concurrently with the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD).
  • the therapeutically effective amounts of the glucocorticoid, antihistamine, and antipyretic are administered concurrently with the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD), the glucocorticoid, antihistamine, and antipyretic may be administered prior to, at the same time as, or after the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered prior to the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered at the same time as the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered after the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered about 1 minute to about 360 minutes, prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD, or any value or range in-between. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 15 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 20 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 25 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 30 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 35 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 40 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 45 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 50 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 55 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 60 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 75 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 90 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 105 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 120 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 135 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 150 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid, antihistamine, and antipyretic are administered 165 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 180 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
  • the glucocorticoid is administered at a certain time prior to the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered about 1 minute to about 240 minutes, prior to the administration of the anti-CD38 antibody, or any value or range in-between. In some embodiments, the glucocorticoid is administered 15 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 20 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 25 minutes prior to the administration of the anti-CD38 antibody.
  • the glucocorticoid is administered 30 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 35 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 40 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 45 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 50 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 55 minutes prior to the administration of the anti-CD38 antibody.
  • the glucocorticoid is administered 60 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 75 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 90 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 105 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 120 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 135 minutes prior to the administration of the anti-CD38 antibody.
  • the glucocorticoid is administered 150 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 165 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 180 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 210 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 240 minutes prior to the administration of the anti-CD38 antibody.
  • the therapeutically effective amount of the anti-CD3 antibody is administered alone. It is to be understood that “alone” does not necessarily connote that the anti- CD3 antibody is administered as a monotherapy, but rather that no other drug is administered simultaneously with, or in the same composition as the anti-CD3 antibody.
  • the anti-CD3 antibody is administered in combination with a substance that reduces the injection volume of a subcutaneous administration of the anti-CD3 antibody.
  • the substance is a recombinant hyaluronidase.
  • the recombinant hyaluronidase is a recombinant human hyaluronidase PH20 (rHuPH20).
  • the hyaluronidase comprises an amino acid sequence of SEQ ID NO: 5.
  • the anti-CD3 antibody is administered in combination with a recombinant hyaluronidase as provided for herein.
  • the anti-CD3 antibody is administered subcutaneously in combination with a recombinant hyaluronidase as provided for herein.
  • the anti-CD3 antibody is administered in combination with a recombinant hyaluronidase comprising an amino acid sequence of SEQ ID NO: 5.
  • the anti-CD3 antibody is administered subcutaneously in combination with a recombinant hyaluronidase comprising an amino acid sequence of SEQ ID NO: 5.
  • the hyaluronidase may be administered at any concentration necessary to achieve the desired effect. In some embodiments, the hyaluronidase is administered at a concentration of about 10,000 U to about 50,000 U, or any value or range in-between. In some embodiments, the hyaluronidase is administered at a concentration of 10,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 20,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 30,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 40,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 50,000 U.
  • Glucocorticoids antihistamines, and antipyretics
  • Glucocorticoids are a class of cortidosteroids which are part of the feedback mechanism of the immune system and act to reduce certain aspects of immune function, such as inflammation.
  • Glucocorticoids are known in the art, and any appropriate glucocorticoid may be used in any of the embodiments provided herein.
  • Non-limiting examples of glucocorticoids include cortisol, cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, and beclometasone.
  • the glucocorticoid is dexamethasone.
  • Antihistamines are a class of drugs that suppress histamine-induced swelling and vasodilation. Antihistamines are known in the art, and any appropriate antihistamine may be used in any of the embodiments provided herein. Non-limiting examples of antihistamines include acrivastine, alimemazine, amitriptyline, amoxapine, azelastine, bilastine, bromodiphenhydramine, bromopheniramine, buclizine, carbinoxamine, cetirizine, chlorodiphenhydramine, chlorphemiramine, chlorpromazine, chlorprothixene, chloropyramine, cinnarizine, clemastine, clomipramine, clozapine, cyclizine, cyproheptadine, desloratadine, dexbrompheniramine, dexchlorpheniramine, dimenhydrinate, dimetindene, diphenhydramine, dosulepin,
  • the antihistamine is selected from the group including, but not limited to, bromodiphenhydramine, chlorodiphenhydramine, diphenhydramine, orphenadrine, or any diphenhydramine equivalent. In some embodiments, the antihistamine is a diphenhydramine equivalent. In some embodiments, the antihistamine is diphenhydramine.
  • Antipyretics are drugs or substances that act to reduce fever. Antipyretics are known in the art, and any appropriate antipyretic may be used in any of the embodiments provided herein. Non-limiting examples of antipyretics include ibuprofen, aspirin, acetaminophen, naproxen, nonsteroidal anti-inflammatory drugs (NSAIDs), salicylates, ketoprofen, flurbiprofen, nimesulide, diclofenac, and celecoxib. In some embodiments, the antipyretic is a NSAID or salicylate.
  • the antipyretic is selected from the group including, but not limited to, ibuprofen, aspirin, acetaminophen, naproxen, ketoprofen, flurbiprofen, nimesulide, diclofenac, and celecoxib.
  • the antipyretic is ibuprofen.
  • the antipyretic is aspirin.
  • the antipyretic is acetaminophen.
  • the antipyretic is naproxen.
  • TABLE 13 - Exemplary Treatment Method a May be elevated to 1500 pg/kg QW or an intermediate dose. b If previously elevated to 1500 pg/kg QW, may be further elevated to 3000 pg/kg Q2W. c May be elevated to 6000 pg/kg Q4W. d Administered with 30,000 units of hyaluronidase.
  • the method comprises a tiered dosing regimen, wherein the teclistamab dose is determined based on a subject weight threshold of 60 kg.
  • the method is as provided in table 14 below:
  • a May be elevated to 300 mg Q4W if Q2W dose determined safe.
  • b May be elevated to 450 mg Q4W if Q2W dose determined safe.
  • c Administered with 30,000 units of hyaluronidase.
  • the methods further comprise a pre-treatment or a co-treatment regimen.
  • the pre-treatment or co-treatment regimen is as provided in table 15 below:
  • the pre-treatment or co-treatment regimen is as provided in table 16 below:
  • the method comprises the treatment regimen of table 13. In some embodiments, the method comprises the treatment regimen of table 14. In some embodiments, the method comprises the treatment regimen of table 13 and further comprises the pre-treatment or co-treatment regimen of table 15. In some embodiments, the method comprises the treatment regimen of table 13 and further comprises the pre-treatment or co-treatment regimen of table 16. In some embodiments, the method comprises the treatment regimen of table 14 and further comprises the pre-treatment or co-treatment regimen of table 15. In some embodiments, the method comprises the treatment regimen of table 14 and further comprises the pre-treatment or co-treatment regimen of table 16.
  • the cancer is a hematological malignancy or a solid tumor.
  • the hematological malignancy is a multiple myeloma, a smoldering multiple myeloma, a monoclonal gammopathy of undetermined significance (MGUS), an acute lymphoblastic leukemia (ALL), a diffuse large B-cell lymphoma (DLBCL), a Burkitt's lymphoma (BL), a follicular lymphoma (FL), a mantle-cell lymphoma (MCL), Waldenstrom’s macroglobulinema, a plasma cell leukemia, a light chain amyloidosis (AL), a precursor B-cell lymphoblastic leukemia, a precursor B-cell lymphoblastic leukemia, an acute myeloid leukemia (AML), a myelodysplastic syndrome (MDS), a chronic lymphocytic leukemia (CLL), a B cell malignancy, a chronic myeloid leukemia (CML), a
  • MUS mantle-
  • the hematological malignancy is multiple myeloma.
  • the multiple myeloma is a newly diagnosed multiple myeloma.
  • the multiple myeloma is a relapsed or a refractory multiple myeloma (RRMM).
  • the multiple myeloma is a high-risk multiple myeloma.
  • Subjects with high-risk multiple myeloma are known to relapse early and have poor prognosis and outcome.
  • Subjects can be classified as having high-risk multiple myeloma is they have one or more of the following cytogenetic abnormalities: t(4;14)(pl6;q32), t(14; 16)(q32;q23), dell7p, IqAmp, t(4; 14)(p 16;q32) and t( 14; 16)(q32;q23), t(4; 14)(pl 6;q32) and dell7p, t( 14; 16)(q32;q23) and dell7p, or t(4; 14)(pl6;q32), t( 14; 16)(q32;q23) and dell7p.
  • the subject having the high-risk multiple myeloma has one or more chromosomal abnormalities comprising: t(4; 14)(pl 6;q32), t( 14; 16)(q32;q23), dell7p, IqAmp, t(4;14)(pl6;q32) and t(14;16)(q32;q23), t(4; 14)(p 16;q32) and dell7p, t(14; 16)(q32;q23) and dell7p; or t(4; 14)(pl 6;q32), t( 14; 16)(q32;q23) and dell7p, or any combination thereof.
  • Symptoms that can be associated are for example a decline or plateau of the well-being of the patient or re-establishment or worsening of various symptoms associated with solid tumors, and/or the spread of cancerous cells in the body from one location to other organs, tissues or cells.
  • the cytogenetic abnormalities can be detected for example by fluorescent in situ hybridization (FISH).
  • FISH fluorescent in situ hybridization
  • an oncogene is translocated to the IgH region on chromosome 14q32, resulting in dysregulation of these genes.
  • t(4; 14)(pl6;q32) involves translocation of fibroblast growth factor receptor 3 (FGFR3) and multiple myeloma SET domain containing protein (MMSET) (also called WHSC1/NSD2)
  • t( 14; 16)(q32;q23) involves translocation of the MAF transcription factor C-MAF.
  • Deletion of 17p (dell7p) involves loss of the p53 gene locus.
  • the multiple myeloma is relapsed or refractory to treatment with the anti-CD38 antibody, lenalidomide, bortezomib, pomalidomide, carfilzomib, elotuzumab, ixazomib, melphalan or thalidomide, or any combination thereof.
  • the multiple myeloma is relapsed or refractory to treatment with the anti-CD38 antibody. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with lenalidomide. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with bortezomib. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with pomalidomide. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with carfilzomib.
  • the multiple myeloma is relapsed or refractory to treatment with elotuzumab. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with ixazomib. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with melphalan. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with or thalidomide.
  • the hematological malignancy is the AML.
  • the AML is AML with at least one genetic abnormality. In some embodiments, the AML is AML with multilineage dysplasia. In some embodiments, the AML is therapy-related AML. In some embodiments, the AML is undifferentiated AML. In some embodiments, the AML is AML with minimal maturation. In some embodiments, the AML is AML with maturation. In some embodiments, the AML is acute myelomonocytic leukemia. In some embodiments, the AML is acute monocytic leukemia. In some embodiments, the AML is acute erythroid leukemia. In some embodiments, the AML is acute megakaryoblastic leukemia. In some embodiments, the AML is acute basophilic leukemia. In some embodiments, the AML is acute panmyelosis with fibrosis. In some embodiments, the AML is myeloid sarcoma.
  • the at least one genetic abnormality is a translocation between chromosomes 8 and 21, a translocation or an inversion in chromosome 16, a translocation between chromosomes 15 and 17, changes in chromosome 11, or mutation in fms-related tyrosine kinase 3 (FLT3), nucleophosmin (NPM1), isocitrate dehydrogenase 1(IDH1), isocitrate dehydrogenase 2 (IDH2), DNA (cytosine-5)-methyltransferase 3 (DNMT3A), CCAAT/enhancer binding protein alpha (CEB PA), U2 small nuclear RNA auxiliary factor 1(U2AF1), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), structural maintenance of chromosomes 1A (SMC1A) or structural maintenance of chromosomes 3 (SMC3).
  • NPM1 nucleophosmin
  • IDH1 isocitrate dehydrogena
  • the at least one genetic abnormality is the translocation between chromosomes 8 and 21. In some embodiments, the at least one genetic abnormality is the translocation or an inversion in chromosome 16. In some embodiments, the at least one genetic abnormality is the translocation between chromosomes 15 and 17. In some embodiments, the at least one genetic abnormality is changes in chromosome 11. In some embodiments, the at least one genetic abnormality is the mutation in fms-related tyrosine kinase 3 (FLT3). In some embodiments, the at least one genetic abnormality is the mutation in nucleophosmin (NPM1).
  • the at least one genetic abnormality is the mutation in isocitrate dehydrogenase 1(IDH1). In some embodiments, the at least one genetic abnormality is the mutation in isocitrate dehydrogenase 2 (IDH2). In some embodiments, the at least one genetic abnormality is the mutation in DNA (cytosine-5)-methyl transferase 3 (DNMT3A). In some embodiments, the at least one genetic abnormality is the mutation in CCAAT/enhancer binding protein alpha (CEBPA). In some embodiments, the at least one genetic abnormality is the mutation in U2 small nuclear RNA auxiliary factor 1(U2AF1).
  • the at least one genetic abnormality is the mutation in enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2). In some embodiments, the at least one genetic abnormality is the mutation in structural maintenance of chromosomes 1A (SMC1A). In some embodiments, the at least one genetic abnormality is the mutation in structural maintenance of chromosomes 3 (SMC3).
  • the at least one genetic abnormality is a translocation t(8; 21)(q22; q22), an inversion inv(16)(pl3; q22), a translocation t( 16; 16)(p 13; q22), a translocation t( 15 ; 17)(q22; ql2), a mutation FLT3-ITD, mutations R132H or R100Q/R104V/F108L/R119Q/I130V in IDH1 or mutations R140Q or R172 in IDH2.
  • the at least one genetic abnormality is the translocation t(8; 21)(q22; q22). In some embodiments, the at least one genetic abnormality is the inversion inv(16)(pl3; q22). In some embodiments, the at least one genetic abnormality is the translocation t( 16; 16)(p 13 ; q22). In some embodiments, the at least one genetic abnormality is the translocation t( 15; 17)(q22; ql2). In some embodiments, the at least one genetic abnormality is the mutation FLT3-ITD. In some embodiments, the at least one genetic abnormality is the mutation R132H in IDH1.
  • the at least one genetic abnormality is the mutation R100Q/R104V/F108L/R119Q/I130V in IDH1. In some embodiments, the at least one genetic abnormality is the mutation R140Q in IDH2. In some embodiments, the at least one genetic abnormality is the mutation R172 in IDH2.
  • the hematological malignancy is the ALL.
  • the ALL is B-cell lineage ALL, T-cell lineage ALL, adult ALL or pediatric ALL.
  • the ALL is B-cell lineage ALL. In some embodiments, the ALL is T-cell lineage ALL. In some embodiments, the ALL is adult ALL. In some embodiments, the ALL is pediatric ALL.
  • the subject with ALL has a Philadelphia chromosome or is resistant or has acquired resistance to treatment with a BCR-ABL kinase inhibitor.
  • the subject with ALL has the Philadelphia chromosome. In some embodiments, the subject with ALL is resistant or has acquired resistance to treatment with a BCR-ABL kinase inhibitor.
  • the Ph chromosome is present in about 20% of adults with ALL and a small percentage of children with ALL and is associated with poor prognosis.
  • patients with Ph-i- positive ALL may be on tyrosine kinase inhibitor (TKI) regimen and may have therefore become resistant to the TKI.
  • the anti-CD38 antibodies may thus be administered to a subject who has become resistant to selective or partially selective BCR-ABL inhibitors.
  • Exemplary BCR-ABL inhibitors are for example imatinib, dasatinib, nilotinib, bosutinib, ponatinib, bafetinib, saracatinib, tozasertib or danusertib.
  • chromosomal rearrangements identified in B-lineage ALL patients are t(v;l lq23) (MLL rearranged), t(l ; 19)(q23;pl3.3); TCF3-PBX1 (E2A-PBX1), t(12;21)(pl3;q22); ETV6- RUNX1 (TEL-AML1) and t(5; 14)(q31 ;q32); IL3-IGH.
  • the subject has ALL with t(v;l lq23) (MLL rearranged), t(l;19)(q23;pl3.3); TCF3-PBX1 (E2A-PBX1), t(12;21)(pl3;q22); ETV6-RUNX1 (TEL-AML1) or t(5; 14)(q31 ;q32); IL3-IGH chromosomal rearrangement.
  • Chromosomal rearrangements can be identified using well known methods, for example fluorescent in situ hybridization, karyotyping, pulsed field gel electrophoresis, or sequencing.
  • the hematological malignancy is the smoldering multiple myeloma. In some embodiments, the hematological malignancy is the MGUS. In some embodiments, the hematological malignancy is the ALL. In some embodiments, the hematological malignancy is the DLBLC. In some embodiments, the hematological malignancy is the BL. In some embodiments, the hematological malignancy is the FL. In some embodiments, the hematological malignancy is the MCL. In some embodiments, the hematological malignancy is Waldenstrom’s macroglobulinema. In some embodiments, the hematological malignancy is the plasma cell leukemia.
  • the hematological malignancy is the AL. In some embodiments, the hematological malignancy is the precursor B- cell lymphoblastic leukemia. In some embodiments, the hematological malignancy is the precursor B-cell lymphoblastic leukemia. In some embodiments, the hematological malignancy is the myelodysplastic syndrome (MDS). In some embodiments, the hematological malignancy is the CLL. In some embodiments, the hematological malignancy is the B cell malignancy. In some embodiments, the hematological malignancy is the CML. In some embodiments, the hematological malignancy is the HCL.
  • MDS myelodysplastic syndrome
  • the hematological malignancy is the blastic plasmacytoid dendritic cell neoplasm. In some embodiments, the hematological malignancy is Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is non-Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is the MZL. In some embodiments, the hematological malignancy is the MALT. In some embodiments, the hematological malignancy is the plasma cell leukemia. In some embodiments, the hematological malignancy is the ALCL. In some embodiments, the hematological malignancy is leukemia. In some embodiments, the hematological malignancy is lymphoma.
  • the subject is an adult 18 years of age or older. In some embodiments, the subject is about 18 to about 100 years old, about 19 to 100 years old, about 20 to 100 years old, about 25 to 100 years old, about 30 to 100 years old, about 35 to 100 years old, about 40 to 100 years old, about 45 to 100 years old, about 50 to 100 years old, about 60 to 100 years old, about 70 to 100 years old, about 80 to 100 years old, about 90 to 100 years old, or any age or age range in-between. In some embodiments, the subject is greater than 100 years old.
  • the subject is about 18 to about 100 years old, about 18 to about 90 years old, about 18 to about 80 years old, about 18 to about 70 years old, about 18 to about 60 years old, about 18 to about 50 years old, about 18 to about 45 years old, about 18 to about 40 years old, about 18 to about 35 years old, about 18 to about 30 years old, about 18 to about 25 years old, about 18 to about 20 years old, about 18 to about 19 years old, or any age or age range inbetween. In some embodiments, the subject is 18 years of age or older.
  • the subject is an adolescent age 12 to 17. In some embodiments, the subject is younger than 18 years old. In some embodiments, the subject is 12. In some embodiments, the subject is 13. In some embodiments, the subject is 14. In some embodiments, the subject is 15. In some embodiments, the subject is 16. In some embodiments, the subject is 16.
  • the subject has an initial diagnosis of multiple myeloma according to the International Myeloma Working Group (IMWG) diagnostic criteria.
  • IMWG International Myeloma Working Group
  • the subject has newly diagnosed multiple myeloma. In some embodiments, the subject has been previously diagnosed with multiple myeloma. In some embodiments, a subject having been previously diagnosed with multiple myeloma has received 1 to 3 prior lines of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 1 prior line of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 2 prior lines of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 3 prior lines of therapy.
  • the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI), immunomodulatory drug (IMiD), or a combination thereof.
  • the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI).
  • the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received an immunomodulatory drug (IMiD).
  • the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI) and an immunomodulatory drug (IMiD).
  • the subject is newly diagnosed with multiple myeloma and is not considered a candidate for high-dose chemotherapy with autologous stem cell transplantation (ASCT).
  • ASCT autologous stem cell transplantation
  • the subject is not considered a candidate for high-dose chemotherapy with ASCT due to advanced age.
  • the subject is not considered a candidate for high-dose chemotherapy with ASCT due to the presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT.
  • the subject is not considered a candidate for high-dose chemotherapy with ASCT due to deferral of high-dose chemotherapy with ASCT as initial treatment.
  • the subject has measurable disease prior to the start of therapy.
  • measurable disease is defined as having a Serum M-protein level > l.Og/dL, urine M-protein level of > 200 mg/24 hours, serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio, or a combination thereof.
  • measurable disease is defined as having a Serum M-protein level > 1.0g/dL.
  • measurable disease is defined as having a urine M-protein level of > 200 mg/24 hours.
  • measurable disease is defined as having a serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio. In some embodiments, measurable disease is defined as having a Serum M-protein level > 1.0g/dL, urine M-protein level of > 200 mg/24 hours, and a serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio.
  • FLC serum IgG free light chain
  • Multiple myeloma is defined as clonal bone marrow plasma cells > 10% or biopsy- proven bony or extramedullary plasmacytoma and at least one of the following two criteria:
  • hypercalcemia is defined as a serum calcium > 0.25 mmol/L (> Img/dL) higher than the upper limit of normal, or > 2.75mmol/L (> 1 Img/dL). In some embodiments, hypercalcemia is defined as a serum calcium > 0.25 mmol/L (> Img/dL) higher than the upper limit of normal. In some embodiments, hypercalcemia is defined as a serum calcium > 2.75mmol/L (> 1 Img/dL).
  • renal insufficiency is defined as creatine clearance ⁇ 40 mL per minute or serum creatine > 177 pmol/L (>2mg/dL). In some embodiments, renal insufficiency is defined as creatine clearance ⁇ 40 mL per minute. In some embodiments, renal insufficiency is defined as serum creatine > 177 pmol/L (>2mg/dL).
  • anemia is defined as a hemoglobin value of > 20 g/L below the lower limit of normal, or a hemoglobin value ⁇ 100 g/L. In some embodiments, anemia is defined as a hemoglobin value of > 20 g/L below the lower limit of normal. In some embodiments, anemia is defined as a hemoglobin value ⁇ 100 g/L. In some embodiments, bone lesions are defined as one or more osteolytic lesions on skeletal radiography, CT, or PET-CT.
  • the subject treated by the methods provided for herein has a partial response (PR) or better. In some embodiments, the subject treated by the methods provided for herein has a very good partial response (VGPR) or better. In some embodiments, the subject treated by the methods provided for herein has a complete response (CR) or better. In some embodiments, the subject treated by the methods provided for herein has a stringent complete response (sCR) or better. Unless indicated otherwise herein, PR, VGPR, CR, sCR, stable disease (SD) and progressive disease (PD) are as defined by the IMWG 2016 criteria. IMWG (2016) response criteria are provided in Table A below.
  • bone marrow criteria for progressive disease are to be used only in participants without measurable disease by M-protein and by FLC levels; “25% increase” refers to M-protein, and FLC, and does not refer to bone lesions, or soft-tissue plasmacytomas and the “lowest response value” does not need to be a confirmed value.
  • CR in such participants indicates a normal FLC ratio of 0.26 to 1.65 in addition to CR criteria listed above.
  • VGPR in such participants requires a >90% decrease in the difference between involved and uninvolved FLC levels.
  • PR is defined as having a greater than 50% reduction of serum M- protein and reduction in 24 hours urinary M-protein by >90% or to ⁇ 200 mg/24 hours.
  • VGPR is defined as having a serum and urine M-protein level detectable by immunofixation but not on electrophoresis or > 90% reduction in serum M-protein plus urine M- protein level ⁇ 100 mg/24 h.
  • CR is defined as having a negative immunofixation on serum and urine and disappearance of any soft tissue plasmacytomas and ⁇
  • sCR is defined as the CR definition as above plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence.
  • the subject treated by the methods provided for herein will have a reduction in BCMA-expressing cells.
  • the subject treated by the methods provided for herein will have an increase in serum or plasma proteins over time, such as but not limited to cytokines (such as IL-6, IFN-y, IL- 10, and IL-2Ra).
  • the subject treated by the methods provided for herein will have pharmacodynamics markers indicative of the mechanism of action of daratumumab, such as reduction in CD38+ NK cells or T reg cells in the periphery.
  • the subject treated by the methods provided for herein will have a reduction in minimal residual disease (MRD) as determined via bone marrow aspirate DNA and next gen sequencing.
  • the subject treated by the methods provided for herein will have a sustained MRD-negative complete response (CR), wherein the subject is MRD-negative for greater than or equal to 12 months.
  • a subject with sustained MRD-negative CR (>12 months) is a subject with CR or better who sustains MRD-negative status, as determined by NGS with sensitivity of 10’ 5 , for at least 12 months without any examination showing MRD positive status or progressive disease in between.
  • the subject treated by the methods provided for herein will have a greater positive response as compared to other lines of therapy.
  • the subject treated by the methods provided for herein will maintain or have an improvement in the European Organization for Research and Treatment of Cancer Quality of Life Questionaire (EORTC-QLQ-C30).
  • the subject treated by the methods provided for herein will maintain or have an improvement in the Patient-reported Outcomeds Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE).
  • the subject treated by the methods provided for herein will maintain or have improvement in the EuroQol Five Dimension Questionnaire 5-Level (EQ-5D- 5L).
  • the subject treated by the methods provided for herein will achieve improvement in one or more symptoms associated with the disease or disorder.
  • the invention also provides a pharmaceutical composition comprising a BCMAxCD3 bispecific antibody and an anti-CD38 antibody as described herein.
  • the composition can comprise a BCMA binding domain comprising a VH of SEQ ID NO: 18 and a VL of SEQ ID NO: 19 and a CD3 binding domain comprising the VH of SEQ ID NO: 20 and the VL of SEQ ID NO: 21, and an anti-CD38 antibody comprising a VH of SEQ ID NO: 34 and the VL of SEQ ID NO:35.
  • the pharmaceutical composition comprises the BCMAxCD3 bispecific antibody comprising the HC1 of SEQ ID NO: 22, the LC1 of SEQ ID NO: 23, the HC2 of SEQ ID NO: 24 the LC2 of SEQ ID NO: 25, and the anti-CD38 antibody comprising the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37.
  • the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises phenylalanine at position 405 and arginine at position 409 in a first heavy chain (HC1) and leucine at position 405 and lysine at position 409 in a second heavy chain (HC2), wherein residue numbering is according to the EU Index.
  • the BCMAxCD3 bispecific antibody further comprises proline at position 228, alanine at position 234 and alanine at position 235 in both the HC1 and the HC2.
  • the disclosure also provides a kit or a combination comprising the BCMAxCD3 bispecific antibody and the anti-CD38 antibody for use in a method of the application.
  • the antibodies used in the methods of the invention binding specific antigens may be selected de novo from, for example, a phage display library, where the phage is engineered to express human immunoglobulins or portions thereof such as Fabs, single chain antibodies (scFv), or unpaired or paired antibody variable regions (Knappik et al., J Mol Biol 296:57-86, 2000;
  • the antibody libraries may be screened for binding to the desired antigen, such as BCM A and the obtained positive clones may be further characterized and the Fabs isolated from the clone lysates, and subsequently cloned as full-length antibodies.
  • desired antigen such as BCM A
  • phage display methods for isolating human antibodies are established in the art. See for example: U.S. Pat. No. 5,223,409; U.S. Pat. No. 5,403,484; U.S. Pat. No. 5,571,698; U.S. Pat. No. 5,427,908; U.S. Pat. No. 5,580,717; U.S. Pat. No. 5,969,108; U.S. Pat. No. 6,172,197; U.S. Pat.
  • T cell redirecting bispecific antibodies may be generated in vitro in a cell-free environment by introducing asymmetrical mutations in the CH3 regions of two monospecific homodimeric antibodies and forming the bispecific heterodimeric antibody from two parent monospecific homodimeric antibodies in reducing conditions to allow disulfide bond isomerization according to methods described in Intl.Pat. Publ. No. WO2011/131746.
  • two monospecific bivalent antibodies are engineered to have certain substitutions at the CH3 domain that promote heterodimer stability; the antibodies are incubated together under reducing conditions sufficient to allow the cysteines in the hinge region to undergo disulfide bond isomerization; thereby generating the bispecific antibody by Fab arm exchange.
  • exemplary reducing agents that may be used are 2- mercaptoethylamine (2-MEA), dithiothreitol (DTT), dithioerythritol (DTE), glutathione, tris(2-carboxyethyl)phosphine (TCEP), L-cysteine and betamercaptoethanol, preferably a reducing agent selected from the group consisting of: 2- mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine.
  • incubation for at least 90 min at a temperature of at least 20°C in the presence of at least 25 mM 2-MEA or in the presence of at least 0.5 mM dithiothreitol at a pH of from 5-8, for example at pH of 7.0 or at pH of 7.4 may be used.
  • CH3 mutations that may be used in a first heavy chain and in a second heavy chain of the bispecific antibody are K409R and/or F405L.
  • Additional CH3 mutations that may be used include technologies such as Duobody® mutations (Genmab), Knob-in-Hole mutations (Genentech), electrostatically-matched mutations (Chugai, Amgen, NovoNordisk, Oncomed), the Strand Exchange Engineered Domain body (SEEDbody) (EMD Serono), and other asymmetric mutations (e.g., Zymeworks).
  • Duobody® mutations are disclosed for example in US9150663 and US2014/0303356 and include mutations F405L/K409R, wild-type/F405L_R409K, T350I_K370T_F405L/K409R, K370W/K409R, D399AFGHILMNRSTVWY/K409R, T366ADEFGHILMQVY/K409R, L368ADEGHNRSTVQ/K409AGRH, D399FHKRQ/K409AGRH, F405IKLSTVW/K409AGRH and Y407LWQ/K409AGRH.
  • Knob-in-hole mutations are disclosed for example in W01996/027011 and include mutations on the interface of CH3 region in which an amino acid with a small side chain (hole) is introduced into the first CH3 region and an amino acid with a large side chain (knob) is introduced into the second CH3 region, resulting in preferential interaction between the first CH3 region and the second CH3 region.
  • Exemplary CH3 region mutations forming a knob and a hole are T366Y/F405A, T366W/F405W, F405W/Y407A, T394W/Y407T, T394S/Y407A, T366W/T394S, F405W/T394S and T366W/T366S_L368A_Y407V.
  • Heavy chain heterodimer formation may be promoted by using electrostatic interactions by substituting positively charged residues on the first CH3 region and negatively charged residues on the second CH3 region as described in US2010/0015133, US2009/0182127, US2010/028637 or US2011/0123532.
  • asymmetric mutations that can be used to promote heavy chain heterodimerization are L351Y_F405A_Y407V/T394W, T366I_K392M_T394W/F405A_Y407V, T366L_K392M_T394W/F405A_Y407V, L351 Y_Y407A/T366A_K409F, L351Y_Y407A/T366V_K409F, Y407A/T366A_K409F, or T350V_L351Y_F405A_Y407V/T350V_T366L_K392L_T394W as described in US2012/0149876 or US2013/0195849.
  • SEEDbody mutations involve substituting select IgG residues with IgA residues to promote heavy chai heterodimerization as described in US20070287170.
  • Additional bispecific or multispecific structures that can be used as BCMAxCD3 bispecific antibodies include Dual Variable Domain Immunoglobulins (DVD) (Int. Pat. Publ. No. WO2009/134776; DVDs are full length antibodies comprising the heavy chain having a structure VH1 -linker- VH2-CH and the light chain having the structure VL1 -linker- VL2-CL; linker being optional), structures that include various dimerization domains to connect the two antibody arms with different specificity, such as leucine zipper or collagen dimerization domains (Int. Pat. Publ. No. WO2012/022811, U.S. Pat. No. 5,932,448; U.S. Pat. No.
  • DVD Dual Variable Domain Immunoglobulins
  • ScFv-, diabody-based, and domain antibodies include but are not limited to, Bispecific T Cell Engager (BiTE) (Micromet), Tandem Diabody (Tandab) (Affimed), Dual Affinity Retargeting Technology (DART) (MacroGenics), Single-chain Diabody (Academic), TCR-like Antibodies (AIT, ReceptorLogics), Human Serum Albumin ScFv Fusion (Merrimack) and COMBODY (Epigen Biotech), dual targeting nanobodies (Ablynx), dual targeting heavy chain only domain antibodies.
  • BiTE Bispecific T Cell Engager
  • Tiandab Tandem Diabody
  • DART Dual Affinity Retargeting Technology
  • AIT TCR-like Antibodies
  • AIT ReceptorLogics
  • Human Serum Albumin ScFv Fusion Merrimack
  • COMBODY Epigen Biotech
  • dual targeting nanobodies Ablynx
  • dual targeting heavy chain only domain antibodies include but are not limited to, Bispecific T
  • the Fc region of the BCMAxCD3 bispecific antibodies such as bispecific or multispecific antibodies or the anti-CD38 antibodies may comprise at least one substitution in the Fc region that reduces binding of the BCMAxCD3 bispecific antibodies to an activating Fey receptor (FcyR) and/or reduces Fc effector functions such as Clq binding, complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) or phagocytosis (ADCP).
  • FcyR activating Fey receptor
  • CDC complement dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • ADCP phagocytosis
  • Fc positions that may be substituted to reduce binding of the Fc to the activating FcyR and subsequently to reduce effector function are substitutions L234A/L235A on IgGl, V234A/G237A/P238S/H268A/V309L/A330S/P331S on IgG2, F234A/L235A on IgG4, S228P/F234A/ L235A on IgG4, N297A on all Ig isotypes, V234A/G237A on IgG2, K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl, H268Q/V309L/ A330S/P331S on IgG2, S267E/L328F on IgGl, L234F/L235E/D265A on IgGl, L2
  • Fc substitutions that may be used to reduce CDC is a K322A substitution.
  • An exemplary wild-type IgGl comprises an amino acid sequence of SEQ ID NO: 16.
  • An exemplary wild-type IgG4 comprises an amino acid sequence of SEQ ID NO: 17.
  • Antibody-dependent cellular cytotoxicity is a mechanism for inducing cell death that depends upon the interaction of antibody-coated target cells with effector cells possessing lytic activity, such as natural killer cells (NK), monocytes, macrophages and neutrophils via Fc gamma receptors (FcyR) expressed on effector cells.
  • effector cells such as natural killer cells (NK), monocytes, macrophages and neutrophils via Fc gamma receptors (FcyR) expressed on effector cells.
  • NK cells express FcyRIIIa
  • monocytes express FcyRI, FcyRII and FcyRIIIa.
  • ADCC activity of the antibodies may be assessed using an in vitro assay using cells expressing the protein the antibody binds to as target cells and NK cells as effector cells.
  • Cytolysis may be detected by the release of label (e.g., radioactive substrates, fluorescent dyes or natural intracellular proteins) from the lysed cells.
  • label e.g., radioactive substrates, fluorescent dyes or natural intracellular proteins
  • target cells are used with a ratio of 1 target cell to 4 effector cells.
  • Target cells are pre-labeled with BATDA and combined with effector cells and the test antibody. The samples are incubated for 2 hours and cell lysis measured by measuring released BATDA into the supernatant. Data is normalized to maximal cytotoxicity with 0.67% Triton X-100 (Sigma Aldrich) and minimal control determined by spontaneous release of BATDA from target cells in the absence of any antibody.
  • ADCP antibody-dependent cellular phagocytosis
  • target cells such as macrophages or dendritic cells
  • ADCP may be evaluated by using monocyte-derived macrophages as effector cells and cells that express the protein the antibody binds to as target cells also engineered to express GFP or another labeled molecule.
  • effector: target cell ratio may be for example 4: 1. Effector cells may be incubated with target cells for 4 hours with or without the antibody of the invention. After incubation, cells may be detached using accutase.
  • Macrophages may be identified with anti-CDl lb and anti-CD14 antibodies coupled to a fluorescent label, and percent phagocytosis may be determined based on % GFP fluorescence in the CD11 + CD14 + macrophages using standard methods.
  • “Complement-dependent cytotoxicity,” or “CDC,” refers to a mechanism for inducing cell death in which the Fc effector domain of a target-bound antibody binds and activates complement component Clq which in turn activates the complement cascade leading to target cell death. Activation of complement may also result in deposition of complement components on the target cell surface that facilitate CDC by binding complement receptors (e.g., CR3) on leukocytes.
  • complement receptors e.g., CR3
  • CDC of cells may be measured for example by plating Daudi cells at IxlO 5 cells/well (50 pE/well) in RPMI-B (RPMI supplemented with 1% BSA), adding 50 pF of test antibodies to the wells at final concentration between 0-100 pg/mE, incubating the reaction for 15 min at room temperature, adding 11 pF of pooled human serum to the wells, and incubation the reaction for 45 min at 37°C. Percentage (%) lysed cells may be detected as % propidium iodide stained cells in FACS assay using standard methods.
  • Binding of the antibody to FcyR or FcRn may be assessed on cells engineered to express each receptor using flow cytometry.
  • 2xl0 5 cells per well are seeded in 96-well plate and blocked in BSA Stain Buffer (BD Biosciences, San Jose, USA) for 30 min at 4°C.
  • Cells are incubated with a test antibody on ice for 1.5 hour at 4°C.
  • R-PE labeled anti -human IgG secondary antibody Jackson Immunoresearch Laboratories
  • the cells are washed twice in stain buffer and then resuspended in 150 pL of Stain Buffer containing 1:200 diluted DRAQ7 live/dead stain (Cell Signaling Technology, Danvers, USA).
  • PE and DRAQ7 signals of the stained cells are detected by Miltenyi MACSQuant flow cytometer (Miltenyi Biotec, Auburn, USA) using B2 and B4 channel, respectively.
  • Live cells are gated on DRAQ7 exclusion and the geometric mean fluorescence signals are determined for at least 10,000 live events collected.
  • FlowJo software (Tree Star) is used for analysis. Data is plotted as the logarithm of antibody concentration versus mean fluorescence signals. Nonlinear regression analysis is performed.
  • Chimeric antigen receptors are genetically engineered receptors. These engineered receptors can be readily inserted into and expressed by immune cells, including T cells in accordance with techniques known in the art. With a CAR, a single receptor can be programmed to both recognize a specific antigen and, when bound to that antigen, activate the immune cell to attack and destroy the cell bearing that antigen. When these antigens exist on tumor cells, an immune cell that expresses the CAR can target and kill the tumor cell.
  • CAR typically comprises an extracellular domain that binds the antigen (e.g., prostate neoantigen), an optional linker, a transmembrane domain, and a cytosolic domain comprising a costimulatory domain and/or a signaling domain.
  • antigen e.g., prostate neoantigen
  • the extracellular domain of CAR may contain any polypeptide that binds the desired antigen (e.g., prostate neoantigen or B cell maturation antigen (BCMA)).
  • the extracellular domain may comprise a scFv, a portion of an antibody or an alternative scaffold.
  • CARs may also be engineered to bind two or more desired antigens that may be arranged in tandem and separated by linker sequences. For example, one or more domain antibodies, scFvs, llama VHH antibodies or other VH only antibody fragments may be organized in tandem via a linker to provide bispecificity or multispecificity to the CAR.
  • the transmembrane domain of CAR may be derived from the transmembrane domain of CD8, an alpha, beta or zeta chain of a T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, 0X40, CD2, CD27, LFA-1 (CDI la, CD 18), ICOS (CD278), 4-1 BB (CD 137), 4-1 BBL, GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRFI), CD 160, CDI 9, IL2R beta, IL2R gamma, IL7R a, ITGA1 , VLA1 , CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CDI Id, IT
  • the intracellular costimulatory domain of CAR may be derived from the intracellular domains of one or more co-stimulatory molecules.
  • Co-stimulatory molecules are well-known cell surface molecules other than antigen receptors or Ec receptors that provide a second signal required for efficient activation and function of T lymphocytes upon binding to antigen.
  • co-stimulatory domains that can be used in CARs are intracellular domains of 4- IBB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 (0X40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM, and ZAP70.
  • the intracellular signaling domain of CAR may be derived from the signaling domains of for example 003 ⁇ , CD3E, CD22, CD79a, CD66d or CD39.
  • “Intracellular signaling domain,” refers to the part of a CAR polypeptide that participates in transducing the message of effective CAR binding to a target antigen into the interior of the immune effector cell to elicit effector cell function, e.g., activation, cytokine production, proliferation and cytotoxic activity, including the release of cytotoxic factors to the CAR-bound target cell, or other cellular responses elicited following antigen binding to the extracellular CAR domain.
  • the optional linker of CAR positioned between the extracellular domain and the transmembrane domain may be a polypeptide of about 2 to 100 amino acids in length.
  • the linker can include or be composed of flexible residues such as glycine and serine so that the adjacent protein domains are free to move relative to one another. Longer linkers may be used when it is desirable to ensure that two adjacent domains do not sterically interfere with one another.
  • Linkers may be cleavable or non-cleavable. Examples of cleavable linkers include 2A linkers (for example T2A), 2A-like linkers or functional equivalents thereof and combinations thereof.
  • the linker may also be derived from a hinge region or portion of the hinge region of any immunoglobulin.
  • Exemplary CARs that may be used are for example CAR that contains an extracellular domain that binds the prostate neoantigen of the invention, CD8 transmembrane domain and CD3 ⁇ signaling domain.
  • Other exemplary CARs contain an extracellular domain that binds the prostate neoantigen of the invention, CD8 or CD28 transmembrane domain, CD28, 41BB or 0X40 costimulatory domain and CD3 ⁇ signaling domain.
  • CARs are generated by standard molecular biology techniques.
  • the extracellular domain that binds the desired antigen may be derived from antibodies or their antigen binding fragments generated using the technologies described herein.
  • the inventors have developed novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
  • ASCT autologous stem cell transplant
  • Newly diagnosed multiple myeloma patients can be broadly categorized as either “transplant eligible” or “transplant ineligible.” Eligibility for transplant is typically based on age, fitness, and comorbidities. Systemic therapy, typically administered as a triplet or quadruplet drug regimen, is the mainstay of treatment both for transplant-eligible and transplant-ineligible patients. It is used in combination with ASCT for those eligible.
  • DRd daratumumab, lenalidomide and dexamethasone
  • VRd bortezomib, lenalidomide and dexamethasone
  • the present inventors have developed novel dosing regimens for BCMAxCD3 bispecific antibodies that provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy.
  • the present inventors have developed novel dosing regimens for combination therapies comprising teclistamab, daratumumab and lenalidomide, that may provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy.
  • Embodiments of the present invention provide novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
  • ASCT autologous stem cell transplant
  • subjects that are newly diagnosed have a diagnosis of multiple myeloma according to the IMWG diagnostic criteria and have not received any prior therapy for multiple myeloma or smoldering myeloma (but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent).
  • RRMM relapsed/refractory multiple myeloma
  • they are not relapsed and/or refractory to prior multiple myeloma therapy but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent).
  • Subjects that are ineligible for ASCT as initial therapy can be ineligible due to (i) advanced age, or (ii) presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT.
  • Subjects that are “not intended” for ASCT as initial therapy are clinically fit enough to undergo ASCT but defer this treatment option.
  • teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy.
  • teclistamab, daratumumab and lenalidomide are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant as initial therapy.
  • Tec-DR improves PFS (progression-free survival) and/or the rate of sustained MRD-negative CR (>12 months) compared with DRd in subjects with newly diagnosed multiple myeloma who are ineligible or not intended for ASCT as initial therapy.
  • teclistamab in combination with daratumumab subcutaneous (SC) and lenalidomide provide an efficacious and safe approach with higher efficacy rates (e.g., mPFS, mOS and MRD negativity rates) compared to the existing standard of care of daratumumab, lenalidomide and dexamethasone (DRd) and/or bortezomib, lenalidomide and dexamethasone (VRd).
  • efficacy rates e.g., mPFS, mOS and MRD negativity rates
  • teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table B below.
  • teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table C below.
  • Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl.
  • teclistamab is administered after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete. It is believed that a DR lead-in cycle and an early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) may reduce the rate of infections compared to the regimen shown in Table B, while still achieving robust efficacy.
  • a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, on a dosing schedule comprising sequential 28-day treatment cycles, wherein administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1 , and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3.
  • the regimen further comprises, in Cycles
  • a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 2, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on Day 1 or
  • (Q4W) dose of 3000 pg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first
  • the regimen further comprises, in Cycles 1-2 only, orally or intravenously administering 20 mg of dexamethasone weekly
  • the method achieves a decrease in new infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
  • the BCMAxCD3 bispecific antibody e.g., QW and/or Q2W
  • the method achieves a decrease in new grade >3 infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
  • the BCMAxCD3 bispecific antibody e.g., QW and/or Q2W
  • the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria.
  • the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria.
  • the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria.
  • the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
  • mPFS median progression-free survival
  • ASCT autologous stem cell transplant
  • the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
  • the method achieves a rate of sustained MRD-negative CR (>12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy.
  • a rate of sustained MRD-negative CR >12 months
  • the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
  • mPFS median progression-free survival
  • the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months.
  • the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
  • mOS median overall survival
  • ASCT autologous stem cell transplant
  • the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years.
  • the method achieves a rate of sustained MRD-negative CR (>12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (>12 months) is greater than a reference rate of sustained MRD- negative CR (>12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
  • ASCT autologous stem cell transplant
  • the rate of sustained MRD- negative CR is greater than the reference rate of sustained MRD-negative CR (>12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%.
  • said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8,
  • a method of treating a cancer in a subject in need thereof comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
  • a BCMA x CD3 bispecific antibody a therapeutically effective amount of an anti-CD38 antibody
  • an immunomodulatory drug IMD
  • a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and
  • a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
  • BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO:
  • a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23 a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • the BCMA x CD3 bispecific antibody is teclistamab.
  • the therapeutically effective amount of the BCMA x CD3 bispecific antibody is selected from 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, 100 mg, 150 mg, 200 mg, 300 mg, and 450 mg.
  • the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
  • the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35. 22. The method of any one of embodiments 1-21, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
  • the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
  • a method of treating a cancer in a subject in need thereof comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x
  • CD3 bispecific antibody of about 60 pg/kg, 240 pg/kg or 300 pg/kg or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg or 6000 pg/kg or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1200 mg to about 2400 mg; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 15 mg to about 50 mg; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the ImiD are administered to the subject for a time sufficient to treat the cancer.
  • a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
  • VH heavy chain variable region
  • HCDRs heavy chain complementarity determining regions
  • LCDRs light chain complementarity determining regions
  • a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
  • a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23 a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • LC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA
  • HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28,
  • the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
  • IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
  • a method of treating a cancer in a subject in need thereof comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of 60 pg/kg, 240 pg/kg, or 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg, or any combination thereof at a frequency selected from, daily, every other day, or weekly; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to
  • a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
  • VH heavy chain variable region
  • HCDRs heavy chain complementarity determining regions
  • LCDRs light chain complementarity determining regions
  • a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
  • a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23 a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • LC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA
  • HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28,
  • the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
  • the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
  • IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
  • a method of treating a cancer in a subject in need thereof comprising: i) determining the subjects weight and assigning a BCMA x CD3 bispecific antibody treatment tier based on said weight, wherein if the subject’s weight is less than or equal to a predetermined threshold the subject is in tier 1, and wherein if the subject’s weight is greater than a pre-determined threshold the subject is in tier 2.
  • the predetermined weight threshold is selected from the group consisting of 50 kg, 55 kg, 60 kg, 65 kg, or 70 kg.
  • a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and
  • a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
  • BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
  • the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
  • the BCMA x CD3 bispecific antibody is teclistamab.
  • anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
  • the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
  • IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
  • the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
  • a method of treating multiple myeloma in a subject in need thereof comprising: i) administering subcutaneously to the subject one or more step-up doses of teclistamab at 60 pg/kg, 240 pg/kg, 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof, on days 2 and 4 of the start of treatment; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of teclistamab of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, 450 mg or any combination thereof, at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of dartumum
  • a method of treating multiple myeloma in a subject in need thereof comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab of 60 pg/kg, on day 2 of a treatment regimen, and administering subcutaneously to the subject a second step-up dose of 240 pg/kg or 300 pg/kg on day 4 of a treatment regimen; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg, on days 8, 15, and 22 of the first treatment cycle; b) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg weekly for each treatment cycle thereafter; wherein the 720 pg/kg weekly dose optionally
  • a method of treating multiple myeloma in a subject in need thereof comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab on day 2 of a first treatment cycle and a second step-up dose of teclistamab on day 4 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the first step-up dose is 3 mg teclistamab and the second step-up dose is 15 mg teclistamab, wherein if the subject weight greater than 60 kg then the first step-up dose is 4 mg teclistamab and the second step-up dose is 24 mg or 25 mg teclistamab; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab, on days 8, 15, and 22 of the first treatment cycle, wherein
  • the pre-treatment regimen comprises: i) intravenously or orally administering a therapeutically effective amount of dexamethasone, wherein the dexamethasone is administered at 20 mg on Day 1 of a first treatment cycle, 16 mg on days 2, 4, 8, 15, and 22 of a first treatment cycle, and 20 mg or 40 mg weekly during each of a second, third, and fourth treatment cycles; ii) intravenously or orally administering a therapeutically effective amount of diphenhydramine, wherein the diphenhydramine is administered at 25 mg to 50 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab; and iii) intravenously or orally administering a therapeutically effective amount of acetaminophen, wherein the acetaminophen is administered at 650 mg to 1000 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab.
  • the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on Day 1 or Day 2), a second step-up dose of 300 pg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 pg/kg (e.g., on Days 8, 15 and 22), in Cycle 2, subcutaneously administering weekly (QW) doses of 1500 pg/kg (e.g., on Days 1, 8, 15 and 22), and starting in Cycle
  • a therapeutically effective combination therapy to the subject comprising the
  • the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on
  • the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, on a dosing schedule comprising sequential 28-day treatment cycles, wherein administration of the daratumumab starts in Cycle 1 , administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3.
  • the BCMAxCD3 bispecific antibody e.g., teclistamab
  • daratumumab e.g., teclistamab
  • daratumumab e.g.,
  • the regimen further comprises: in Cycles 1 and 2 only, orally or intravenously administering dexamethasone to the subject. 130.
  • mPFS median progression-free survival
  • a CR or better i.e., a sCR or a CR
  • response criteria in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
  • ASCT autologous stem cell transplant
  • mPFS median progression-free survival
  • ASCT autologous stem cell transplant
  • mOS median overall survival
  • ASCT autologous stem cell transplant
  • Embodiment #125 examples are described in Table 19 and Table 26.
  • the BCMAxCD3 bispecific antibody e.g., teclistamab
  • daratumumab are administered starting from Cycle 1
  • lenalidomide is not started until Cycle 2
  • dexamethasone may be administered in Cycles 2-4.
  • the BCMAxCD3 bispecific antibody e.g., teclistamab
  • Cycle 2 and is administered monthly (Q4W) starting from its second treatment cycle, i.e., Cycle 3 of the regimen
  • daratumumab and lenalidomide are started in Cycle 1 (i.e., there is a DR lead-in in Cycle 1) and dexamethasone may be administered in Cycles 1-2.
  • JNJ-957 comprises a BCMA binding arm BCMB69 comprising a HC and LC of SEQ ID NOs 22 and 23, respectively, and a CD3 binding arm CD3B219 comprising a HC and a LC of SEQ ID NOs 24 and 25, respectively.
  • the amino acid sequences of the CDRs, VH, VL, HC, and LC of each binding arm of teclistamab are shown in Tables 7a and 7b in the detailed description section of this disclosure.
  • PBMCs Peripheral blood mononuclear cells
  • BM-MNCs bone marrow mononuclear cells
  • BM-localized MM cells were identified and analysed for cell surface marker expression levels by staining l.OxlO 6 cells/mL with HuMax-003 (CD38) FITC (this antibody binds to an epitope distinct from the epitope bound by daratumumab, Janssen Pharmaceuticals), CD138 PE, CD56 PC7, CD45 Krome Orange (all Beckman Coulter), CD269 (BCMA) APC (Biolegend), CD274 (PD-L1) BV421 and CD19 APC-H7 (both Becton Dickinson).
  • BM or PB immune cell subsets were identified and analysed for cell surface marker expression levels by staining l.OxlO 6 cells/mL with CD45 Krome Orange, CD56 PC7 (both Beckman Coulter), CD 14 APC- H7, CD 19 APC-H7, CD3 V450, CD4 APC-H7 or PE, CD8 FITC, CD45-RA APC, CD 127 PE.Cy7, CD62L PE, CD274 (PD-1) BV421, CD 16 APC, HLA-DR APC-H7 (all Becton Dickinson) and CD25 PE (Dako) or with CD4 BUV395 (BD Biosciences), CD8 BUV737 (BD Biosciences), PD-1 BV421 (BD Biosciences), TIM-3 BV650 (BD Biosciences), CD3 BV711 (BD Biosciences), CD45RO BV786 (BD Biosciences), CD38 Humab-003-FITC (Janssen), CD45RA PerCP
  • Flow cytometry was performed using a 7-laser LSRFORTESSA (Becton Dickinson). Fluorescent labeled beads (CS&T beads, Becton Dickinson) were used daily to monitor the performance of the flow cytometer and verify optical path and stream flow. This procedure enables controlled standardized results and allows the determination of long-term drifts and incidental changes within the flow cytometer. No changes were observed which could affect the results. Compensation beads were used to determine spectral overlap, compensation was automatically calculated using Diva software. Flow cytometry data were analyzed using FACS Diva software.
  • % lysis 1- (mean BLI signal in the presence of effector cells and JNJ-957 / mean BLI signal in the presence of effector cells in untreated wells) xl00%.
  • the LUC-transduced MM cell line 4 was also co-cultured with PB MNCs, obtained from MM patients before initiation of daratumumab monotherapy and at the time of best response to daratumumab monotherapy (effector to target ratio of 9:1).
  • the BLI assay was performed as described before.
  • Cytogenetic abnormalities were assessed in purified MM cells by fluorescence in situ hybridization (FISH) and single nucleotide polymorphism (SNP) array. High-risk disease was defined by the presence of del(17p), del(lp), ampl(lq), t(4; 14) or t( 14; 16) 2 .
  • Soluble BCMA was measured in cell culture supernatants using MSD GOLDTM 96-well Small Spot Streptavidin SECTOR plates (Meso Scale Diagnostics), according to the manufacturer’s recommended protocol.
  • Cytokines [interferon-gamma (IFN-y), interleukin (IL)-2, IL-6, IL-8, IL- 10, and tumor necrosis factor-alpha (TNF-a)] in the cell culture supernatants were analyzed using V-Plex proinflammatory Panel 1 Human Kit (Meso Scale Diagnostics), according to the manufacturer’s protocol.
  • % expected lysis (% lysis with JNJ-957 + % lysis with daratumumab) - (% lysis with JNJ-957 x % lysis with daratumumab), as described before.
  • the null hypothesis of “additive effects” was rejected, if the observed values were significantly higher (P ⁇ 0.05) than the expected values.
  • Treatment Regimens A, B, C, D, F which included i) teclistamab (SC), daratumumab (SC), and pomalidomide (oral); ii) teclistamab (SC), daratumumab (SC), lenalidomide (oral), and bortezomib (SC) for a 21 day treatment cycle; iii) teclistamab (SC) and nirogacestat (SC); iv) teclistamab (SC) and lenalidomide (oral); and v) teclistamab (SC), daratumumab (SC), lenalidomide (oral), and bortezomib (SC) for a 28 day treatment cycle.
  • the overall aim of the study was to characterize the safety and tolerability of teclistamab combination regimens by assessing the incidence and severity of adverse events (AEs), laboratory values, and the frequency and type of dose-limiting toxicities (DLTs). Safety was monitored throughout the study by a Study Evaluation Team (SET).
  • AEs adverse events
  • DLTs dose-limiting toxicities
  • Treatment Regimen B teclistamab + daratumumab + lenalidomide + bortezomib (tec- dara-len-bor)
  • Treatment Regimen F teclistamab + daratumumab + lenalidomide + bortezomib; 28- day cycle schedule
  • teclistamab is administered at a starting dose of 720 pg/kg SC weekly, 1 dose level below the monotherapy recommended phase 2 dose (RP2D) (i.e., 1500 pg/kg SC weekly) as determined based on preliminary data from Study 64007957MMY1001 (NCT03145181).
  • R2D monotherapy recommended phase 2 dose
  • ICF informed consent form
  • Serum M-protein level > 1.0 g/dL; or b. Urine M-protein level > 200 mg/24 hours; or c. Light chain multiple myeloma; Serum Ig free light chain (FLC) > 10 mg/dL and abnormal serum Ig kappa lambda FLC ratio.
  • FLC Serum Ig free light chain
  • ANC absolute neutrophil count
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • G-CSF granulocyte colony stimulating factor
  • GM-CSF granulocyte-macrophage colonystimulating factor
  • peg-G-CSF pegylated granulocyte colony-stimulating factor
  • RBC red blood cell
  • hormonal contraception e.g., oral estrogen/progestin
  • a male or female condom with or without spermicide e.g., spermicidal foam/gel/cream/suppository
  • Women of childbearing potential must be on 2 methods of reliable birth control simultaneously while receiving treatment and until 100 days after last dose of treatment: one highly effective form of contraception (tubal ligation, intrauterine device, hormonal [oral, injectable, transdermal patches, vaginal rings, or implants], or partner’s vasectomy), and 1 additional effective contraceptive method (male latex or synthetic condom, diaphragm, or cervical cap) iv. Agree to pregnancy testing (serum or urine) within 100 days after the last dose of study treatment.
  • one highly effective form of contraception tubal ligation, intrauterine device, hormonal [oral, injectable, transdermal patches, vaginal rings, or implants], or partner’s vasectomy
  • 1 additional effective contraceptive method male latex or synthetic condom, diaphragm, or cervical cap
  • Prior antitumor therapy as follows, in the specified time frame prior to the first dose of the study treatment: a. Targeted therapy, epigenetic therapy, or treatment with an investigational drug or an invasive investigational medical device within 21 days or at least 5 half-lives, whichever is less. b. Gene-modified adoptive cell therapy (e.g., CAR modified T cells, NK cells) within 3 months. c. mAh treatment for multiple myeloma treatment within 21 days. d. Cytotoxic therapy within 21 days. e. PI therapy within 14 days. f. Immunomodulatory agent therapy within 7 days. g. Radiotherapy within 14 days. However, if palliative focal radiation is used, the participant is eligible irrespective of the end date of radiotherapy.
  • a. Targeted therapy, epigenetic therapy, or treatment with an investigational drug or an invasive investigational medical device within 21 days or at least 5 half-lives, whichever is less.
  • Gene-modified adoptive cell therapy e.g., CAR
  • Active central nervous system involvement or exhibition of clinical signs of meningeal involvement of multiple myeloma. If either is suspected, brain magnetic resonance imaging (MRI) and lumbar cytology are required.
  • Active plasma cell leukemia > 2.0 x 10 9 /L plasma cells by standard differential
  • Waldenstrom s macroglobulinemia
  • POEMS syndrome polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes
  • HBV hepatitis B surface antigen
  • Participants with resolved infection i.e., participants who are HBsAg negative but positive for hepatitis B core antibody [anti-HBc] and/or positive for hepatitis B surface antibody [anti-HBs]
  • PCR real-time polymerase chain reaction
  • HCV hepatitis C virus
  • Any serious underlying medical condition such as: a. Evidence of serious active viral, bacterial, or uncontrolled systemic fungal infection. b. Active autoimmune disease or a documented history of autoimmune disease, with the exception of vitiligo, resolved childhood atopic dermatitis, and prior Graves’ disease that is currently euthyroid based on clinical symptoms and laboratory testing. c. Disabling psychiatric conditions (e.g., ongoing alcohol or drug abuse), sever dementia, or altered mental status. d.
  • SC step up doses of 60 pg/kg and 240 pg/kg were administered on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 720 pg/kg administered on days 8, 15, and 22 of treatment cycle 1.
  • Teclistamab 720 pg/kg SC was administered weekly for the second and all subsequent treatment cycles.
  • a separate cohort was administered teclistamab at 1500 pg/kg SC on days 8, 15, and 22 of treatment cycle 1, followed by 1500 pg/kg SC weekly for treatment cycle 2 and then 4000 pg/kg SC biweekly or 3000 pg/kg SC biweekly for treatment cycle 3 and each subsequent treatment cycle.
  • Dosing of teclistamab SC optionally can be further modified as determined by the SET.
  • teclistamab SC 720 pg/kg QW may be elevated to 1500 pg/kg QW as determined by the SET, and may be further elevated to, for example, 3000 pg/kg Q2W beginning at treatment cycle 3 and 6000 pg/kg Q4W or 3000 pg/kg Q4W beginning at treatment cycle 7.
  • Subjects are stratified into two cohorts based on weight. Subjects ⁇ 60 kg receive a teclistamab SC step up doses of 3 mg and 15 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 100 mg on days 8, 15, and 22 of treatment cycle 1. Teclistamab SC 100 mg is administered weekly for the second treatment cycle, followed by 200 mg Q2W starting at treatment cycle 3 and, if Q2W dosing is deemed safe, 300 mg Q4W starting at treatment cycle 7. Subjects > 60 kg receive a teclistamab SC step up doses of 4 mg and 25 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 150 mg on days 8, 15, and 22 of treatment cycle 1. Tec SC 150 mg is administered weekly for the second treatment cycle, followed by 300 mg Q2W starting at treatment cycle 3 and, if Q2W dosing is deemed safe, 450 mg Q4W starting at treatment cycle 7.
  • Daratumumab was administered to all subjects by SC injection at a dose of 1800 mg with 30000 units hyaluronidase to decrease injection volume required.
  • Daratumumab SC was administered weekly in treatment cycles 1 and 2, Q2W in treatment cycles 3-6, and Q4W thereafter.
  • Lenalidomide was administered orally at 25 mg every day for 21 days of a 28-day treatment cycle. Lenalidomide treatment started at treatment cycle 2.
  • daratumumab and teclistamab were administered on the same day, daratumumab was administered first.
  • Step-up dose 1 of teclistamab is administered at least 20 hours after daratumumab SC.
  • the first treatment dose of teclistamab is administered 3 hours after daratumumab.
  • Subsequent treatment doses of teclistamab are administered 1 hour after daratumumab SC.
  • the treatment regimen also includes administration of dexamethasone weekly during treatment cycles 2, 3, and 4.
  • Dexamethasone may be administered at 40 mg, or if the participant is greater than 75 years of age or less than or equal to 75 years of age and has a BMI less than 18.5 dexamethasone may be administered at a dose of 20 mg.
  • the treatment also includes required pretreatment medications.
  • the required pretreatment medications include dexamethasone (glucocorticoid), IV or oral.
  • Dexamethasone is administered at 20 mg on day 1 for the first dose of daratumumab SC and 16 mg for all step-up doses and first treatment doses of teclistamab SC.
  • Dexamethasone is administered on day 1 for the first dose of daratumumab SC and after grade 2 administration related reaction (sAAR) and Grade > 3 sARRs related to daratumumab SC. Dexamethasone is also administered after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC. Dexamthasone is administered approximately 1 to 3 hours prior to daratumumab SC or teclistamab SC on days on which daratumumab SC is not administered. Additional required pretreatment medications include diphenhydramine (anti-histamine) or an equivalent and acetaminophen (antipyretic).
  • Diphenhydramine is administered at 25 mg to 50 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC, and after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC.
  • Diphenhydramine is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC.
  • Acetaminophen is administered at 650 mg to 1000 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC, and after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC.
  • Acetaminophen is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC.
  • pretreatment medications should be given prior to daratumumab SC. If there are greater than 4 hours between administration of study drugs, a repeat dose of dexamethasone 8 mg must be given prior to the bispecific antibody.
  • CRS was frequent, but low-grade. All CRS events were grade 1/2, and 95% of the events occurred during cycle 1 treatment doses. Median time to onset was 2 days (range, 1-8) and median duration was 2 days (range, 1-22). A summary of CRS events is provided in Table 22 below.
  • ASTCT American Society for Transplantation and Cellular Therapy
  • CRS cytokine release syndrome
  • dara daratumumab
  • len lenalidomide
  • tec teclistamab.
  • Infections were common, but generally low grade. Infections of any grade occurred in 24 patients (75%). The most common infections were CO VID- 19, pneumonia, and upper respiratory infection. Grade 3/4 infections occurred in 9 patients (28.1%). 3 patients (9.4%) had CO VID-19 pneumonia. 1 patient (3.1%) discontinued due to an AE (COVID-19), considered unrelated to the study drugs. This patient subsequently died due to COVID-19. There were no other deaths related to AEs.
  • the rate of infections for the teclistamab + daratumumab + lenalidomide treatment regimen was similar to teclistamab monotherapy, but the rate of grade 3/4 infections was lower than the monotherapy.
  • a summary of the infection data is provided in Table 23 below.
  • CR complete response
  • dara daratumumab
  • IMWG International Myeloma Working Group
  • len lenalidomide
  • ORR overall response rate
  • PR partial response
  • sCR stringent complete response
  • tec teclistamab
  • VGPR very good partial response
  • the overall response rate (ORR) was 13/13 evaluable patients (median follow-up, 8.61 mo) at 0.72 mg/kg and 13/16 evaluable patients (median follow-up was less mature at 4.17 mo) at 1.5 mg/kg. Very good partial response or better was achieved in 12 patients at the 0.72 mg/kg dose and was not mature for the 1.5 mg/kg group. Median time to first response was 1.0 mo (range, 0.7-2.0). Preliminary pharmacokinetic concentrations of tec in combination with dara-len were similar to those seen with tec monotherapy.
  • Tec-dara-len was well tolerated, with a safety profile consistent with tec or dara-len individually. Promising ORR supports the potential for this combination to have enhanced early disease control through the addition of teclistamab.
  • the randomized phase 3 MajesTEC-7 study described in Example 2 will compare tec-dara-len vs the combination of dara, len, and dexamethasone in patients with NDMM ineligible or not intended for autologous stem cell transplant as initial treatment.
  • EXAMPLE 2 A Phase 3 Randomized Study Comparing Teclistamab in Combination with Daratumumab SC and Lenalidomide (Tec-DR) versus Daratumumab SC, Lenalidomide, and Dexamethasone (DRd), and Talquetamab in Combination with Daratumumab SC and Lenalidomide (Tal-DR) versus DRd, in Patients with Newly Diagnoses Multiple Myeloma Who are Either Ineligible or not Intended for Autologous Stem Cell Transplant as Initial Therapy (MajesTEC-7)
  • the overall aim is to compare the efficacy between Tec-DR and DRd, and Tal-DR and DRd, in terms of PFS (progression-free survival, which refers to duration from the date of randomization to either progressive disease or death, whichever comes first) and minimal residual disease (MRD)-negative complete response (CR).
  • PFS progression-free survival, which refers to duration from the date of randomization to either progressive disease or death, whichever comes first
  • MRD minimal residual disease
  • Table 25 Objectives and endpoints of the phase 3 study.
  • ASCT autologous stem cell transplant
  • Tec-DR cohorts varying doses of Teclistamab (Tec) are administered with a constant dose of daratumumab (dara), lenalidomide (len), and dexamethasone.
  • DRd cohorts constant doses of daratumumab (dara), lenalidomide (len), and dexamethasone are administered.
  • teclistamab fixed dosing subjects are stratified into two cohorts based on weight. Subjects ⁇ 60 kg receive a teclistamab SC step up doses of 3 mg and 15 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 100 mg on days 8, 15, and 22 of treatment cycle 1. Teclistamab SC 100 mg is administered weekly for the second treatment cycle, followed by 200 mg Q2W starting at treatment cycle 3, and 200 mg Q4W starting at treatment cycle 7. Subjects > 60 kg receive a teclistamab SC step up doses of 4 mg and 25 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 150 mg on days 8, 15, and 22 of treatment cycle 1. Tec SC 150 mg is administered weekly for the second treatment cycle, followed by 300 mg Q2W starting at treatment cycle 3, and 300 mg Q4W starting at treatment cycle 7.
  • Daratumumab is administered to all subjects by SC injection at a dose of 1800 mg with 30000 units hyaluronidase to decrease injection volume required.
  • Daratumumab SC is administered weekly in treatment cycles 1 and 2, Q2W in treatment cycles 3-6, and Q4W thereafter.
  • Lenalidomide is administered orally at 25 mg every day for 21 days of a 28-day treatment cycle. Lenalidomide treatment started at treatment cycle 2.
  • dexamethasone is administered orally or intravenously at 20 mg weekly.
  • dexamethasone is administered during treatment cycles 2, 3, and 4.
  • dexamethasone is administered at 40 mg, or if the participant is greater than 75 years of age or has a BMI less than 18.5 dexamethasone is administered at a dose of 20 mg.
  • dexamethasone is administered weekly for all cycles.
  • daratumumab and teclistamab were administered on the same day, daratumumab was administered first.
  • Step-up dose 1 of teclistamab is administered at least 20 hours after daratumumab SC.
  • the first treatment dose of teclistamab is administered 3 hours after daratumumab.
  • Subsequent treatment doses of teclistamab are administered at least 15 minutes after daratumumab SC.
  • the treatment also includes required and optional pretreatment medications.
  • the required pretreatment medications include dexamethasone (glucocorticoid), IV or oral.
  • Dexamethasone is administered at 16 mg on days 1, 2, 4 and 8 of treatment cycle 1.
  • Dexamthasone is administered approximately 1 to 3 hours prior to daratumumab SC or teclistamab SC on days on which daratumumab SC is not administered.
  • Additional required pretreatment medications include diphenhydramine (anti-histamine) or an equivalent and acetaminophen (antipyretic).
  • Diphenhydramine is administered at 25 mg to 50 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC.
  • Diphenhydramine is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC.
  • Acetaminophen is administered at 650 mg to 1000 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC.
  • Acetaminophen is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC.
  • montelukast (10 mg) may be administered at the discretion of the examiner prior to administration of daratumumab.
  • pretreatment medications should be given prior to daratumumab SC.
  • dexamethasone 8 mg must be given prior to the bispecific antibody.
  • a participant treated with Tec-DR who experiences Grade >2 CRS/sARR related to teclistamab or daratumumab SC must receive dexamethasone 16 mg, diphenhydramine 25 to 50 mg or equivalent, and acetaminophen 650 to 1000 mg as pretreatment medication for at least the subsequent dose of the study drug to which the event was related.
  • These event-driven pretreatment medications are not required if already being administered as planned pretreatment described above.
  • Dexamethasone doses scheduled as background treatment for Cycles 2-4 in participants treated with Tec-DR can be used as event-driven pretreatment medication.
  • Eligible patients will be aged >18 years with NDMM and are either ineligible or not intended for ASCT as initial therapy, have measurable disease, and an ECOG performance status score 0-2.
  • a safety run-in phase for Tec-DR will precede the randomized phase.
  • Approximately 1000 patients will be randomized 1: 1 to receive Tec-DR or DRd.
  • Treatment will continue until progressive disease, death, intolerable toxicity, loss to follow-up, withdrawal of consent, or end of study, whichever occurs first.
  • the dual primary endpoints are PFS and minimal residual disease (MRD)-negative complete response (CR) sustained for at least 12 months.
  • Secondary endpoints include rate of CR or better, overall survival, rate of MRD-negative CR, rate of very good partial response or better, PFS2, patient-reported outcomes, and adverse events (AEs). Response will be assessed using 2016 International Myeloma Working Group criteria. AEs will be graded by Common Terminology Criteria for AEs v5.0, except for cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, which will be graded by American Society for Transplantation and Cellular Therapy guidelines. Results from this trial will provide insights into a possible new treatment regimen (Tec-DR) for patients with NDMM, which may provide improved outcomes.
  • Tec-DR Treatment regimen
  • Tec-DR SRI 1 The study treatment that was administered for the Tec-DR Safety Run-In Cohort 1 (Tec- DR SRI 1) is provided in Table 26. Participants enrolled in Safety Run-in Cohort 1 were initially treated with fixed dose teclistamab in combination with DR. Following implementation of a protocol amendment, due to a safety measure, all participants treated with fixed dose teclistamab were transitioned to weight-based teclistamab.
  • IV intravenous
  • PO per os (oral);
  • Q2W every other week;
  • SC subcutaneous
  • IV intravenous
  • PO per os (oral);
  • Q2W every other week;
  • SC subcutaneous
  • dexamethasone may be administered at a dose of 20 mg.
  • the rate of any grade infection was 92.3%
  • the rate of Grade 3+ infections was 38.5%
  • the rate of Grade 3 or 4 infections was 34.6%.
  • Tec-DR SRI 2 The study treatment to be administered for the Tec-DR Safety Run-In Cohort 2 (Tec-DR SRI 2), and the Randomized Tec-DR Arm, is provided in Table 29.
  • the aim of the dosing regimen used in Tec-DR SRI 2 is to achieve robust efficacy while improving the safety profile seen in Tec-DR SRI 1, including reduced rates of infections.
  • Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl.
  • teclistamab will be given after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete.
  • the early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) was implemented with the aim of reducing the rate of severe infections.
  • the teclistamab SC dose schedule for Safety Run-in Cohort 2 of the study will include 2 step-up doses (0.06 and 0.3 mg/kg) on Days 1 and 3 of Cycle 2 followed by 1.5 mg/kg on Days 8 and 15 of Cycle 2 and 3 mg/kg Q4W starting from Cycle 3.
  • modified Q4W regimen will resemble the exposure (concentration-time profile) of the 0.72 mg/kg QW dose, which has shown robust efficacy and good safety in combination with daratumumab and lenalidomide in MajesTEC-2 (Study 64007957MMY1004).

Landscapes

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

Abstract

Disclosed herein are methods of treating cancers with combinations of a BCMAxCD3 bispecific antibody, an anti-CD3 antibody, and an immunomodulatory drug. Also provided are pharmaceutical compositions comprising the same and methods for generating the antibodies of the present disclosure.

Description

METHODS OF TREATING CANCERS
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[0001] The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on October 4, 2024, is named
“258199061102_JBI6761WOPCTl_Sequence_Listing.xml” and is 65 kilobytes in size.
CROSS REFERENCE TO RELATED APPLICATIONS
[0002] This application claims the benefit of priority of U.S. Provisional Patent Application No. 63/382,089, filed on November 2, 2022, which is incorporated by reference herein in its entirety.
FIELD OF THE INVENTION
[0003] Disclosed are methods of treating cancers with BCMA x CD3 bispecific antibodies and combinations comprising BCMA x CD3 bispecific antibodies, anti-CD38 antibodies, and immunomodulatory drugs.
BACKGROUND OF THE INVENTION
[0004] Multiple myeloma (MM) is a cancer of the plasma cells. Mechanistically, multiple myeloma is characterized by production of monoclonal proteins (M-proteins) comprised of pathological immunoglobulins or fragments of such, which have lost their function. The proliferation of multiple myeloma cells leads to subsequent displacement from the normal bone marrow niche, while overproduction of M-proteins causes characteristic osteolytic lesions, increased susceptibility to infections, hypercalcemia, renal insufficiency or failure, and neurological complications.
[0005] Treatment options for multiple myeloma have improved over time and vary depending on the aggressiveness of the disease, underlying prognostic factors, physical condition of the patient, and existing comorbidities. Therapeutic options include proteasome inhibitors (Pls), immunomodulatory drugs (IMiDs), alkylating agents, monoclonal antibodies (mAbs), antibody drug conjugate, histone deacetylase inhibitor, nuclear protein export inhibitor, chimeric antigen receptor (CAR) T cell therapy and stem cell transplantation.
[0006] Despite these therapeutic achievements, the disease recurs and is associated with additional risk factors (e.g., comorbidities or increasing age), thus warranting the need for novel therapeutic approaches, such as new dosage and treatment regimens. In particular in the elderly population, for which stem cell transplantation is often not a viable option, and in patients with refractory disease who exhausted all available therapies, multiple myeloma remains an incurable malignancy and an unmet medical need with significant morbidity and mortality.
SUMMARY OF THE INVENTION
[0007] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD3 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
[0008] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
[0009] In some embodiments, the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
[0010] In some embodiments, the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen-binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
[0011] In some embodiments, the BCMA x CD3 bispecific antibody is teclistamab.
In some embodiments, the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
[0012] In some embodiments, the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
[0013] In some embodiments, the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
[0014] In some embodiments, the anti-CD38 antibody is daratumumab.
[0015] In some embodiments, the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
[0016] In some embodiments, the IMiD is lenalidomide.
[0017] In some embodiments, the method further comprises administering a pre-treatment to the subject. [0018] In some embodiments, the pre-treatment comprises administration of a glucocorticoid, an antihistamine, an antipyretic, or a combination thereof.
[0019] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of about 60 pg/kg, 240 pg/kg or 300 pg/kg or of about 3 mg, 4 mg, 15 mg, 24 mg or 25 mg; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg or 6000 pg/kg or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1200 mg to about 2400 mg; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 15 mg to about 50 mg; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
[0020] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of 60 pg/kg, 240 pg/kg, or 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg or 25 mg, or any combination thereof at a frequency selected from, daily, every other day, or weekly; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 25 mg at a frequency of daily or weekly; wherein the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
[0021] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) determining the subjects weight and assigning a BCMA x CD3 bispecific antibody treatment tier based on said weight, wherein if the subject’s weight is less than or equal to a pre-determined threshold the subject is in tier 1, and wherein if the subject’s weight is greater than a pre-determined threshold the subject is in tier 2; ii) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody; iii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody; iv) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody; and v) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD); wherein the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
[0022] In some embodiments, the predetermined weight threshold is selected from the group consisting of 50 kg, 55 kg, 60 kg, 65 kg, or 70 kg.
[0023] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) administering subcutaneously to the subject one or more step-up doses of teclistamab at 60 pg/kg, 240 pg/kg, 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof, on days 2 and 4 of the start of treatment; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of teclistamab of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, 450 mg or any combination thereof, at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of dartumumab of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of lenalidomide of about 25 mg at a frequency of once per day; wherein teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
[0024] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) administering subcutaneously to the subject a first step-up dose of teclistamab of 60 pg/kg, on day 2 of a treatment regimen, and administering subcutaneously to the subject a second step-up dose of 240 pg/kg or 300 pg/kg on day 4 of a treatment regimen; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising a) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg, on days 8, 15, and 22 of the first treatment cycle; b) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg weekly for each treatment cycle thereafter, wherein wherein the 720 pg/kg weekly dose optionally is elevated to 1500 pg/kg weekly, wherein wherein the 1500 pg/kg weekly dose optionally is elevated to 3000 pg/kg biweekly starting at a third treatment cycle, and wherein the 3000 pg/kg biweekly dose optionally is elevated to 6000 pg/kg once every four weeks starting at a seventh treatment cycle; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; and wherein teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
[0025] In some embodiments, a method of treating cancer in a subject in need thereof is provided, the method comprising i) administering subcutaneously to the subject a first step-up dose of teclistamab on day 2 of a first treatment cycle and a second step-up dose of teclistamab on day 4 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the first step-up dose is 3 mg teclistamab and the second step-up dose is 15 mg teclistamab, and wherein if the subject weight greater than 60 kg then the first step-up dose is 4 mg teclistamab and the second step-up dose is 24 mg or 25 mg teclistamab; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising a) administering subcutaneously to the subject a treatment dose of teclistamab, on days 8, 15, and 22 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; b) administering subcutaneously to the subject a treatment dose of teclistamab, weekly for a second treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; c) administering subcutaneously to the subject a treatment dose of teclistamab, biweekly for a third, fourth, fifth, and sixth treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 300 mg; and d) administering subcutaneously to the subject a treatment dose of teclistamab, once every four weeks for a seventh and subsequent treatment cycles, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, optionally wherein the treatment dose is elevated to 300 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 300 mg, optionally wherein the treatment dose is elevated to 450 mg; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; and wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
[0026] In some embodiments, the methods provided for herein further comprise administering a pre-treatment regimen. In some embodiments, the pre-treatment regimen comprises i) intravenously or orally administering a therapeutically effective amount of dexamethasone, wherein the dexamethasone is administered at 20 mg on Day 1 of a first treatment cycle, 16 mg on days 2, 4, 8, 15, and 22 of a first treatment cycle, and 20 mg or 40 mg weekly during each of a second, third, and fourth treatment cycles; ii) intravenously or orally administering a therapeutically effective amount of diphenhydramine, wherein the diphenhydramine is administered at 25 mg to 50 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab; and iii) intravenously or orally administering a therapeutically effective amount of acetaminophen, wherein the acetaminophen is administered at 650 mg to 1000 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab. In some embodiments, the pre-treatment regimen further comprises administration of montelukast at 10 mg.
BRIEF DESCRIPTION OF THE DRAWINGS
[0027] The foregoing summary, as well as the following detailed description of the invention, will be better understood when read in conjunction with the appended drawings. It should be understood that the invention is not limited to the precise embodiments shown in the drawings.
[0028] FIG. 1 is a schematic overview of the Phase 1 study of teclistamab administered in combination with daratumumab and lenalidomide, as of Nov. 2022 (MajesTEC-2 Regimen E; NCT04722146).
[0029] FIG. 2 illustrates the interleukin 6 profile for cohorts treated with teclistamab, daratumumab, and lenalidomide, as of Nov. 2022 (MajesTEC-2 Regimen E; NCT04722146).
[0030] FIG. 3 is a schematic overview of the initial Phase 3 study design for teclistamab administered in combination with daratumumab and lenalidomide (teclistamab-DR) as compared to daratumumab and lenalidomide in the absence of teclistamab, as of Nov. 2022 (MajesTEC-7; NCT05552222).
[0031] FIG. 4 is a schematic overview of the updated Phase 3 study design that includes 3 arms, Teclistamab-DR, Talquetamab-DR and DRd, as of Oct. 2023 (MajesTEC-7; NCT05552222).
DETAILED DESCRIPTION OF THE INVENTION
[0032] The disclosed methods may be understood more readily by reference to the following detailed description taken in connection with the accompanying figures, which form a part of this disclosure. It is to be understood that the disclosed methods are not limited to the specific methods described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of the claimed methods. All patents, patent applications and publications cited herein are incorporated by reference as if set forth fully herein. [0033] Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein.
[0034] As used herein, the singular forms “a,” “an,” and “the” include the plural unless the context clearly dictates otherwise. Thus, for example, reference to “a cell” includes a combination of two or more cells, and the like.
[0035] Unless otherwise stated, any numerical value, such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term “about.” Thus, a numerical value typically includes ± 10% of the recited value. For example, a dosage of 10 mg includes 9 mg to 11 mg. As used herein, the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.
[0036] “About” when used in reference to numerical ranges, cutoffs, or specific values means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. Unless explicitly stated otherwise within the Examples or elsewhere in the Specification in the context of an assay, result or embodiment, “about” means within one standard deviation per the practice in the art, or a range of up to 10%, whichever is larger.
[0037] “Antibodies” is meant in a broad sense and includes immunoglobulin molecules including monoclonal antibodies including murine, human, humanized and chimeric monoclonal antibodies, antigen binding fragments, multispecific antibodies, such as bispecific, trispecific, tetraspecific etc., dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding site of the required specificity. “Full length antibodies” are comprised of two heavy chains (HC) and two light chains (LC) inter-connected by disulfide bonds as well as multimers thereof (e.g., IgM). Each heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region (comprised of domains CHI, hinge, CH2 and CH3). Each light chain is comprised of a light chain variable region (VL) and a light chain constant region (CL). The VH and the VL regions may be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with framework regions (ER). Each VH and VL is composed of three CDRs and four FR segments, arranged from amino-to-carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. Immunoglobulins may be assigned to five major classes, IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified as the isotypes IgAl, IgA2, IgGl, IgG2, IgG3 and IgG4. Antibody light chains of any vertebrate species may be assigned to one of two clearly distinct types, namely kappa (K) and lambda ( ), based on the amino acid sequences of their constant domains.
[0038] “Antigen binding fragment” or “antigen binding domain” refers to a portion of an immunoglobulin molecule that binds an antigen. Antigen binding fragments may be synthetic, enzymatically obtainable or genetically engineered polypeptides and include the VH, the VL, the VH and the VL, Fab, F(ab')2, Fd and Fv fragments, domain antibodies (dAb) consisting of one VH domain or one VL domain, shark variable IgNAR domains, camelized VH domains, minimal recognition units consisting of the amino acid residues that mimic the CDRs of an antibody, such as FR3-CDR3-FR4 portions, the HCDR1, the HCDR2 and/or the HCDR3 and the LCDR1, the LCDR2 and/or the LCDR3. VH and VL domains may be linked together via a synthetic linker to form various types of single chain antibody designs where the VH/VL domains may pair intramolecularly, or intermolecularly in those cases when the VH and VL domains are expressed by separate single chain antibody constructs, to form a monovalent antigen binding site, such as single chain Fv (scFv) or diabody; described for example in Int. Patent Publ. Nos.
W01998/44001, WO1988/01649, WO1994/13804 and W01992/01047.
[0039] “BCMA” refers to human B-cell maturation antigen, also known as CD269 or TNFRSF17. A human BCMA (UniProt Q02223) contains the amino acid sequence of SEQ ID NO: 1. The extracellular domain of the human BCMA encompasses residues 1-54 of SEQ ID NO: 1.
[0040] “Bispecific” refers to an antibody that specifically binds two distinct antigens or two distinct epitopes within the same antigen. The bispecific antibody may have cross-reactivity to other related antigens, for example to the same antigen from other species (homologs), such as human or monkey, for example Macaca cynomolgus (cynomolgus, cyno) or Pan troglodytes, or may bind an epitope that is shared between two or more distinct antigens.
[0041] “Cancer” refers to a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth results in the formation of malignant tumors that invade neighboring tissues and may also metastasize to distant parts of the body through the lymphatic system or bloodstream. A “cancer” or “cancer tissue” can include a tumor.
[0042] “CD3” refers to a human antigen which is expressed on T cells as part of the multimolecular T cell receptor (TCR) complex and which consists of a homodimer or heterodimer formed from the association of two or four receptor chains: CD3 epsilon, CD3 delta, CD3 zeta and CD3 gamma. Human CD3 epsilon comprises the amino acid sequence of SEQ ID NO: 2. SEQ ID NO: 3 shows the extracellular domain of CD3 epsilon.
[0043] “CD38” refers to the CD38 protein (synonyms: ADP-ribosyl cyclase 1, cADPr hydrolase 1, cyclic ADP-ribose hydrolase 1). A human CD38 (UniProt accession no. P28907) has the amino acid sequence as shown in SEQ ID NO: 4. CD38 is a single pass type II transmembrane protein with amino acid residues 1-21 representing the cytosolic domain, amino acid residues 22- 42 representing the transmembrane domain, and residues 43-300 representing the extracellular domain.
[0044] “CH3 region” or “CH3 domain” refers to the CH3 region of an immunoglobulin. The CH3 region of human IgGl antibody corresponds to amino acid residues 341-446. However, the CH3 region may also be any of the other antibody isotypes as described herein.
[0045] “Chimeric antigen receptor” or “CAR” refers to engineered T cell receptors which graft a ligand or antigen specificity onto T cells (for example naive T cells central memory T cells effector memory T cells or combinations thereof). CARs are also known as artificial T- cell receptors, chimeric T-cell receptors or chimeric immunoreceptors. CARs comprise an extracellular domain capable of binding to an antigen, a transmembrane domain and at least one intracellular domain. CAR intracellular domain comprises a polypeptide known to function as a domain that transmits a signal to cause activation or inhibition of a biological process in a cell. The transmembrane domain comprises any peptide or polypeptide known to span the cell membrane and that can function to link the extracellular and signaling domains. A chimeric antigen receptor may optionally comprise a hinge domain which serves as a linker between the extracellular and transmembrane domains.
[0046] “Combination” means that two or more therapeutics are administered to a subject together in a mixture, concurrently as single agents or sequentially as single agents in any order. A “combination therapy” may refer to a therapeutically effective regimen that comprises administration of two or more anti-multiple myeloma agents to a subject to treat multiple myeloma.
[0047] “Complementarity determining regions” (CDR) are antibody regions that bind an antigen. CDRs may be defined using various delineations such as Kabat (Wu et al. J Exp Med 132: 211- 50, 1970) (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991), Chothia (Chothia et al. J Mol Biol 196: 901-17, 1987), IMGT (Lefranc et al. Dev Comp Immunol 27: 55-77, 2003) and AbM (Martin and Thornton J Bmol Biol 263: 800-15, 1996). The correspondence between the various delineations and variable region numbering are described (see e.g., Lefranc et al. Dev Comp Immunol 27: 55-77, 2003; Honegger and Pluckthun, J Mol Biol 309:657-70, 2001; International ImMunoGeneTics (IMGT) database; Web resources, http://www_imgt_org). Available programs such as abY sis by UCL Business PLC may be used to delineate CDRs. The term “CDR”, “HCDR1”, “HCDR2”, “HCDR3”, “LCDR1”, “LCDR2” and “LCDR3” as used herein includes CDRs defined by any of the methods described supra, Kabat, Chothia, IMGT or AbM, unless otherwise explicitly stated in the specification. Correspondence between the numbering system, including, for example, the Kabat numbering and the IMGT unique numbering system, is well known to one skilled in the art (see, e.g., Kabat; Chothia; Martin; Lefranc et al.).
[0048] Table 1. Kabat, IMGT, AbM, and Chothia numbering systems.
Figure imgf000014_0001
[0049] “Comprising” is intended to include examples encompassed by the terms “consisting essentially of’ and “consisting of;” similarly, the term “consisting essentially of’ is intended to include examples encompassed by the term “consisting of.” Unless the context clearly requires otherwise, throughout the description and the claims, the words “comprise,” “comprising,” and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of “including, but not limited to.”
[0050] “Enhance” or “enhanced” refers to enhancement in one or more functions of a test molecule when compared to a control molecule or a combination of test molecules when compared to one or more control molecules. Exemplary functions that can be measured are tumor cell killing, T cell activation, relative or absolute T cell number, Fc-mediated effector function (e.g., ADCC, CDC and/or ADCP) or binding to an Fey receptor (FcyR) or FcRn. “Enhanced” may be an enhancement of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more, or a statistically significant enhancement.
[0051] “Fc gamma receptor” (FcyR) refers to well-known FcyRI, FcyRIIa, FcyRIIb or FcyRIII. Activating FcyR includes FcyRI, FcyRIIa and FcyRIII.
[0052] “Fixed dose,” also referred to as “flat dose,” refers to a dose that is administered to a subject without correction for the subject’s specific body weight or body surface area. A fixed dose, sometimes referred to as a flat dose, is therefore provided as an absolute amount of the agent (e.g., mg drug), and not as a weight-based amount (e.g. pg/kg or pg drug per kg body weight). For example, a subject weighing 65kg may be administered the same flat dose in milligrams as a subject weighing 85kg. A flat dose may be administered according to a predefined class or category of body weight, but is not modified according to the subject’s specific weight. For example, a “Flat Dose A” may be administered if a patient is greater than a predefined threshold weight (e.g., > 60 kg), whereas a different “Flat Dose B” may be administered if the patent is less than or equal to the pre-defined threshold weight (e.g., < 60 kg).
[0053] As used herein, “weight-based” refers to administration of a dose amount that is based on the subject’s specific body weight; for example, 3 mg/kg refers to a dose of 3 milligrams of antibody per kilogram of the subject’s body weight. Unless otherwise specified herein, when a dose is described in a unit of “mg/kg” or “pg/kg,” weight-based dosing is being employed.
[0054] Unless otherwise specified herein, a BCMAxCD3 bispecific antibody, such as teclistamab, is administered on a dosing schedule based on sequential 28-day treatment cycles, for example, Cycle 1 starts on Day 1 of Cycle 1 and ends on Day 28 of Cycle 1, and then Day 1 of Cycle 2 starts the day after Day 28 of Cycle 1 and ends on Day 28 of Cycle 2, and then Day 1 of Cycle 3 starts the day after Day 28 of Cycle 2 and ends on Day 28 of Cycle 3, and so on. As used herein with respect to treatment cycles, “Cl” refers to Cycle 1, “C2” refers to Cycle 2, “C3” refers to Cycle 3, and so on. Multiple cycles may also be described, e.g., “C3-6” refers to Cycles 3-6 (Cycles 3, 4, 5 and 6). A cycle number with a “+” symbol refers to that cycle and all subsequent cycles, e.g., “C3+” refers to from Cycle 3 and all subsequent cycles (i.e., C3, C4, C5, C6, C7, and so on).
[0055] According to certain embodiments, administration of the BCMAxCD3 bispecific antibody begins in Cycle 1 of a therapeutically effective regimen. According to other embodiments, in which the BCMAxCD3 antibody is administered as part of a combination therapy with one or more additional anti-multiple myeloma agents, administration of the BCMAxCD3 bispecific antibody may start in Cycle 2 or later, i.e., administration of one or more other anti-multiple myeloma agents (but not the BCMAxCD3 bispecific antibody) may start in Cycle 1 and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2 or later. Thus, in accordance with such embodiments, the first treatment cycle that includes administration of the BCMAxCD3 bispecific antibody is Cycle 2 or later.
[0056] As used herein “Q4W” means once every four weeks (i.e., once every 28 days), “Q2W” (also referred to as “bi-weekly” or “biweekly”) means once every two weeks, and “QW” (also referred to as “weekly”) means once weekly. Q4W is also referred to herein as “monthly.” The terms “Q4W” and “monthly” are used interchangeably to refer to once every 4 weeks or once every 28 days (e.g., in sequential 28-day cycles, a first treatment dose occurs on Day 1 of Cycle 1, a second treatment dose occurs on Day 1 of Cycle 2, etc.). Administration of a treatment dose once weekly (QW) is also referred to herein as a weekly dosing schedule; for example, a 28-day treatment cycle may have a weekly dosing schedule that comprises four doses one week apart from each other (e.g., on Days 1, 8, 15 and 22), or three doses one week apart from each other (e.g., on Days 8, 15 and 22), or two doses one week apart from each other (e.g., on Days 8 and 15). Administration of a treatment dose once every two weeks (Q2W) is also referred to herein as a bi-weekly dosing schedule. Administration of a treatment dose once every four weeks (Q4W) is also referred to herein as a monthly dosing schedule. Dosing regimens may be described herein in terms of the dose amount and frequency; for example, “Cl: 1500 pg/kg QW” refers to administration of 1500 pg/kg once per week in Cycle 1 of a therapeutically effective regimen, “C3-6: 3000 pg/kg Q2W” refers to administration of 3000 pg/kg once every two weeks from Cycle 3 through Cycle 6, “C3+: 3000 pg/kg Q4W” refers to administration of 3000 pg/kg once every four weeks starting in Cycle 3, etc.
[0057] Additional abbreviations used herein include the following: CR, complete response; PR, partial response; Q2W, once every 2 weeks; Q4W, once every 4 weeks; QW, once weekly; RP2D, recommended phase 2 dose; SUD, step-up dose.
[0058] “Human antibody” refers to an antibody that is optimized to have minimal immune response when administered to a human subject. Variable regions of human antibody are derived from human immunoglobulin sequences. If human antibody contains a constant region or a portion of the constant region, the constant region is also derived from human immunoglobulin sequences. Human antibody comprises heavy and light chain variable regions that are “derived from” sequences of human origin if the variable regions of the human antibody are obtained from a system that uses human germline immunoglobulin or rearranged immunoglobulin genes. Such exemplary systems are human immunoglobulin gene libraries displayed on phage, and transgenic non-human animals such as mice or rats carrying human immunoglobulin loci. “Human antibody” typically contains amino acid differences when compared to the immunoglobulins expressed in humans due to differences between the systems used to obtain the human antibody and human immunoglobulin loci, introduction of somatic mutations or intentional introduction of substitutions into the frameworks or CDRs, or both. Typically, “human antibody” is at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical in amino acid sequence to an amino acid sequence encoded by human germline immunoglobulin or rearranged immunoglobulin genes. In some cases, “human antibody” may contain consensus framework sequences derived from human framework sequence analyses, for example as described in Knappik et al., (2000) J Mol Biol 296:57-86, or synthetic HCDR3 incorporated into human immunoglobulin gene libraries displayed on phage, for example as described in Shi et al., (2010) J Mol Biol 397:385-96, and in Int. Patent Publ. No. W02009/085462. Antibodies in which at least one CDR is derived from a non-human species are not included in the definition of “human antibody”.
[0059] “Humanized antibody” refers to an antibody in which at least one CDR is derived from non-human species and at least one framework is derived from human immunoglobulin sequences. Humanized antibody may include substitutions in the frameworks so that the frameworks may not be exact copies of expressed human immunoglobulin or human immunoglobulin germline gene sequences.
[0060] As used herein, IMWG diagnostic criteria for multiple myeloma have been met when the participant was diagnosed. IMWG diagnostic criteria are known in the art. Multiple myeloma may be defined as clonal BMPCs >10% or biopsy-proven bony or extramedullary plasmacytoma3 and at least 1 of the following:
• Evidence of end-organ damage, specifically:
- C: Hypercalcemia: serum calcium >0.25 mmol/L (>1 mg/dL) higher than the ULN or >2.75 mmol/L (>11 mg/dL)
- R: Renal insufficiency: creatinine clearance <40 mL per minb or serum creatinine >177 pmol/L (>2 mg/dL)
- A: Anemia: hemoglobin value of >20 g/L below the lower limit of normal, or a hemoglobin value <100 g/L
- B: Bone lesions: 1 or more osteolytic lesions on skeletal radiography, CT, or PET-CTc d
• Any 1 or more of the following biomarkers of malignancy:
- Clonal BMPC%a >60%
- Involvedmninvolved sELC ratio6 >100
- >1 focal lesions on MRI studiesf a. Clonality should be established by showing K/A-I ight-chain restriction on flow cytometry, immunohistochemistry, or immunofluorescence. Bone marrow plasma cell percentage should preferably be estimated from a core biopsy specimen; in case of a disparity between the aspirate and core biopsy, the highest value should be used. b. Measured or estimated by validated equations. c. If bone marrow has less than 10% clonal plasma cells, more than one bone lesion is required to distinguish from solitary plasmacytoma with minimal marrow involvement. d. PET-CT=18F-fluorodeoxyglucose PET with CT. e. These values are based on the serum Freelite assay (The Binding Site Group, Birmingham, UK). The involved FLC must be >100 mg/L.f. Each focal lesion must be 5 mm or more in size.
[0061] “Isolated” refers to a homogenous population of molecules (such as synthetic polynucleotides or a protein such as an antibody) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step. “Isolated antibody” refers to an antibody that is substantially free of other cellular material and/or chemicals and encompasses antibodies that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity.
[0062] “Monoclonal antibody” refers to an antibody obtained from a substantially homogenous population of antibody molecules, i.e., the individual antibodies comprising the population are identical except for possible well-known alterations such as removal of C-terminal lysine from the antibody heavy chain or post-translational modifications such as amino acid isomerization or deamidation, methionine oxidation or asparagine or glutamine deamidation. Monoclonal antibodies typically bind one antigenic epitope. A bispecific monoclonal antibody binds two distinct antigenic epitopes. Monoclonal antibodies may have heterogeneous glycosylation within the antibody population. Monoclonal antibody may be monospecific or multispecific such as bispecific, monovalent, bivalent or multivalent.
[0063] “Mutation” refers to an engineered or naturally occurring alteration in a polypeptide or polynucleotide sequence when compared to a reference sequence. The alteration may be a substitution, insertion or deletion of one or more amino acids or polynucleotides. [0064] “Non-fixed combination” refers to separate pharmaceutical compositions of the BCMAxCD3 bispecific antibody and the anti-CD38 antibody administered as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the subject.
[0065] “Multispecific” refers to an antibody that specifically binds at least two distinct antigens or at least two distinct epitopes within the same antigen. Multispecific antibody may bind for example two, three, four or five distinct antigens or distinct epitopes within the same antigen.
[0066] “Pharmaceutical composition” refers to composition that comprises an active ingredient and a pharmaceutically acceptable carrier.
[0067] “Pharmaceutically acceptable carrier” or “excipient” refers to an ingredient in a pharmaceutical composition, other than the active ingredient, which is nontoxic to a subject.
[0068] “Philadelphia chromosome” or “Ph” refers to a well-known chromosomal translocation between chromosomes 9 and 22, resulting in the oncogenic BCR-ABL gene fusion with constitutively active tyrosine kinase activity. The translocation results in a portion of the BCR gene from chromosome 22ql 1 becoming fused with a portion of the ABL gene from chromosome 9q34, and is designated as t(9;22)(q34;ql 1) under the International System for Human Cytogenetic Nomenclature (ISCN). Depending on the precise location of the fusion, the molecular weight of the resulting fusion protein can range from 185 to 210 kDa. “Philadelphia chromosome” refers to all BCR-ABL fusion proteins formed due the (9;22)(q34;ql 1) translocation.
[0069] “Recombinant” refers to DNA, antibodies and other proteins that are prepared, expressed, created or isolated by recombinant means when segments from different sources are joined to produce recombinant DNA, antibodies or proteins.
[0070] “Reduce” or “reduced” refers to a reduction in one or more functions of a test molecule when compared to a control molecule or a combination of test molecules when compared to one or more control molecules. Exemplary functions that can be measured are tumor cell killing, T cell activation, relative or absolute T cell number, Fc-mediated effector function (e.g., ADCC, CDC and/or ADCP) or binding to an Fey receptor (FcyR) or FcRn. “Reduced” may be a reduction of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more, or a statistically significant enhancement.
[0071] “rHuPh20” refers to recombinant human hyalurodinase having the amino acid sequence of SEQ ID NO: 5, which is a recombinant hyaluronidase (HYLENEX® recombinant) described in Int'l Pat. Pub. No. W02004/078140.
[0072] “Refractory to a therapy” refers to a cancer that is not amendable to surgical intervention and is initially unresponsive to the therapy.
[0073] “Relapsed” refers to a cancer that responded to a treatment but then returns.
[0074] “Step-up dose” - refers to a dose of an active agent that is administered to a subject prior to a treatment dose. A step-up dose is lower than the treatment dose. To prevent or lessen certain toxicities, such as cytokine release syndrome (CRS), a “priming” dose strategy may include one or more lower step-up dose(s) followed by higher treatment doses.
[0075] A “step-up phase” refers to an initial phase of a therapeutically effective regimen in which at least one step-up dose of a therapeutic is administered to the subject. A step-up phase may also include one or more treatment doses, i.e., a step-up phase may include one or more step-up doses followed by one or more treatment doses; for example, a step-up phase may include two step-up doses followed by two treatment doses. In particular embodiments, the step- up phase is 28 days, i.e., the step-up phase is a 28-day cycle of a therapeutically effective regimen.
[0076] “Subject” includes any human or nonhuman animal. “Nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Except when noted, the terms “patient” or “subject” are used interchangeably.
[0077] “BCMAxCD3 bispecific antibody” refers to a molecule containing two or more binding regions, wherein one of the binding regions specifically binds the cell surface antigen B-cell maturation antigen (BCMA) on a target cell or tissue and wherein a second binding region of the molecule specifically binds a T cell antigen CD3. This dual/multi-target binding ability recruits T cells to the target cell or tissue leading to the eradication of the target cell or tissue. [0078] “Therapeutically effective amount” refers to an amount effective, at doses and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount may vary depending on factors such as the disease state, age, sex, and weight of the individual, and the ability of a therapeutic or a combination of therapeutics to elicit a desired response in the individual. Exemplary indicators of an effective therapeutic or combination of therapeutics that include, for example, improved well-being of the patient.
[0079] “Treat” or “treatment” refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder. Beneficial or desired clinical results include alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if a subject was not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
[0080] “Treatment dose” refers to a dose of the active agent that is administered to a subject to treat a disease. A treatment dose may be administered at a regular dosing interval on a repetitive basis (e.g. weekly, biweekly, monthly). A treatment dose may be preceded by one or more step- up doses.
[0081] “Tumor cell” or a “cancer cell” refers to a cancerous, pre-cancerous or transformed cell, either in vivo, ex vivo, or in tissue culture, that has spontaneous or induced phenotypic changes. These changes do not necessarily involve the uptake of new genetic material. Although transformation may arise from infection with a transforming virus and incorporation of new genomic nucleic acid, uptake of exogenous nucleic acid or it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene. Transformation/cancer is exemplified by morphological changes, immortalization of cells, aberrant growth control, foci formation, proliferation, malignancy, modulation of tumor specific marker levels, invasiveness, tumor growth in suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo. [0082] The numbering of amino acid residues in the antibody constant region throughout the specification is according to the EU index as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991), unless otherwise explicitly stated. Antibody constant chain numbering can be found for example at ImMunoGeneTics website, at IMGT Web resources at IMGT Scientific charts.
[0083] The substitutions in the CH3 region are expressed as modified position(s) in the first CH3 domain of the first heavy chain/ modified position(s) in the second CH3 domain of the second heavy chain. For example, F405L/K409R refers to a F405L mutation in the first CH3 region and K09R mutation in the second CH3 region. L351Y_F405A_Y407V/T394W refers to L351Y, F40FA and Y407V mutations in the first CH3 region and T394W mutation in the second CH3 region. D399FHKRQ/K409AGRH refers to mutation in which D399 may be replaced by F, H, K R or Q, and K409 may be replaced by A, G, R or H.
[0084] Conventional one and three-letter amino acid codes are used herein as shown in Table 2.
[0085] Table 2. Amino acid abbreviations.
Figure imgf000023_0001
| Valine _ | Vai _ | V _ |
[0086] In one general aspect, the disclosure relates to a method of treating a cancer in a subject in need thereof. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody, an effective amount of an anti-CD38 antibody, and an effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. The method may comprise administering any one of a BCMA x CD3 bispecific antibody, an anti-CD38 antibody, and an IMid, or any combination of a BCMA x CD3 bispecific antibody, an anti-CD38 antibody, and an IMid. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an anti-CD38 antibody to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an IMid to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody and an effective amount of an anti-CD38 antibody to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of an anti-CD38 antibody and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises administering an effective amount of a BCMA x CD3 bispecific antibody, an effective amount of an anti-CD38 antibody, and an effective amount of an IMiD to the subject for a time sufficient to treat the cancer.
BCMA x CD3 bispecific antibodies
[0087] B-cell maturation antigen (BCMA) is a cell membrane bound tumor necrosis factor receptor family member involved in differentiation of B-cells to plasma cells. Expression of BCMA is restricted to the B-cell lineage where it is predominantly expressed in the interfollicular region of germinal centers and on differentiated plasma cells and plasmablasts. BCMA is virtually absent on naive and memory B cells. A BCMA x CD3 bispecific antibody targets the CD3 receptor complex on T cells and BCMA on plasma cells. The dual binding site allows the bispecific antibody to draw CD3+ T cells in close proximity to myeloma cells, without regard to T cell receptor specificity or reliance on MHC Class 1 molecules on the surface of antigen presenting cells for activation, leading to cell death of the BCMA-positive cell.
[0088] Any suitable BCMA x CD3 bispecific antibody can be used in any of the embodiments provided herein. Exemplary multispecific and/or bispecific formats include dual targeting molecules including, but not limited to, Dual Targeting (DT)-Ig (GSK/Domantis), Two-in-one Antibody (Genetech) and mAb2 (F-Star), Dual Variable Domain (DVD)-Ig (Abbott), Ts2Ab (Medlmmune/AZ) and BsAb (Zymogenetics), HERCULES (Biogen Idee) and TvAb (Roche), ScFv/Fc Fusions (Academic Institution), SCORPION (Emergent BioSolutions/Trubion, Zymogenetics/BMS) and Dual Affinity Retargeting Technology (Fc-DART) (MacroGenics), F(ab)2 (Megarex/AMGEN), Dual-Action or Bis-Fab (Genetech), Dock-and-Lock (DNL) (ImmunoMedics), Bivalent Bispecific (Biotecnol) and Fab-Fv (UCB-Celltech), Bispecific T Cell Engager (BITE) (Micromet), Tandem Biabody (Tandab) (Affimend), Dual Affinity Retargeting Technology (DART) (MarcroGenics), Single-chain Diabody (Academic), TCR-like Antibodies (AIT, ReceptorLogics), Human Serum Albumin ScFv Fusion (Merrimack) and COMBODY (Epigen Biotech), dual targeting nanobodies (Ablynx), and dual targeting heavy chain only domain antibodies. Various formats of bispecific antibodies have been described, for example in Chames and Baty (2009) Curr Opin Drug Disc Dev 12: 276 and in Nunez-Prado et al., (2015) Drug Discovery Today 20(5):588-594
[0089] In some embodiments, the BCMA x CD3 bispecific antibody is an antigen binding fragment. Exemplary antigen binding fragments are Fab, F(ab')2, Fd and Fv fragments.
[0090] In some embodiments, the BCMA x CD3 bispecific antibody is chimeric, humanized or human.
[0091] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 6, the HCDR2 of SEQ ID NO: 7, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 12, the HCDR2 of SEQ ID NO: 13, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17. The HCDRs and LCDRs of the BCMA x
CD3 bispecific antibody are recited in Table 3 below:
TABLE 3: Exemplary CDRs of BCMA x CD3 bispecific antibody
Figure imgf000026_0001
[0092] The CDRs recited in the table above are of the Kabat number system. However, as provided for herein, the CDRs of the present disclosure may be provided by any appropriate numbering system, such as any of the Kabat, Chothia, IMGT or AbM numbering systems. Tables 4-6 below provides exemplary CDRs utilizing the Chothia, AbM, and IMGT numbering systems:
TABLE 4: Exemplary CDRs of BCMA x CD3 bispecific antibody - Chothia numbering system
Figure imgf000026_0002
Figure imgf000027_0001
TABLE 5: Exemplary CDRs of BCMA x CD3 bispecific antibody - AbM numbering system
Figure imgf000027_0002
TABLE 6: Exemplary CDRs of BCMA x CD3 bispecific antibody - IMGT numbering system
Figure imgf000027_0003
Figure imgf000028_0001
[0093] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 46, the HCDR2 of SEQ ID NO: 47, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 48, the HCDR2 of SEQ ID NO: 49, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17.
[0094] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 50, the HCDR2 of SEQ ID NO: 51, the HCDR3 of SEQ ID NO: 8, and a VL having the LCDR1 of SEQ ID NO: 9, the LCDR2 of SEQ ID NO: 10 and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 52, the HCDR2 of SEQ ID NO: 53, the HCDR3 of SEQ ID NO: 14, and a VL having the LCDR1 of SEQ ID NO: 15, the LCDR2 of SEQ ID NO: 16 and the LCDR3 of SEQ ID NO: 17.
[0095] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising a VH having the HCDR1 of SEQ ID NO: 54, the HCDR2 of SEQ ID NO: 55, the HCDR3 of SEQ ID NO: 56, and a VL having the LCDR1 of SEQ ID NO: 57, a LCDR2 comprising the amino acid sequence DD, and the LCDR3 of SEQ ID NO: 11, and a CD3 binding domain comprising a VH having the HCDR1 of SEQ ID NO: 58, the HCDR2 of SEQ ID NO: 59, the HCDR3 of SEQ ID NO: 60, and a VL having the LCDR1 of SEQ ID NO: 61, a LCDR2 comprising the amino acid sequence GT and the LCDR3 of SEQ ID NO: 17.
[0096] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain comprising the VH of SEQ ID NO: 18 and the VL of SEQ ID NO: 19, and a CD3 binding domain comprising the VH of SEQ ID NO: 20 and the VL of SEQ ID NO: 21.
[0097] In some embodiments, the BCMA x CD3 bispecific antibody that binds BCMA comprises a first heavy chain (HC1) of SEQ ID NO: 22, a first light chain (LC1) of SEQ ID NO: 23, a second heavy chain (HC2) of SEQ ID NO: 24, and a second light chain (LC2) of SEQ ID NO: 25.
[0098] In some embodiments, the BCMA binding arm of the BCMS x CD3 bispecific antibody and the CD3 binging arm of the BCMA x CD3 bispecific antibody comprise the amino acid sequences as provided in Tables 7a and 7b.
TABLE 7a: Sequences of the BCMA binding arm of a BCMA x CD3 bispecific antibody.
Figure imgf000029_0001
Figure imgf000030_0001
TABLE 7b: Sequences of the CD3 binding arm of a BCMA x CD3 bispecific antibody.
Figure imgf000030_0002
Figure imgf000031_0001
[0099] In some embodiments, the BCMA x CD3 bispecific antibody comprises a BCMA binding domain that binds BCMA selected from the group consisting of the BCMA binding domain of ACTR cancer therapy by Seattle Genetics, AFM-26, ALLO-715, anti-BCMA allogenic CAR-T cell therapy by CRISPR Therapeutics, anti-BCMA CAR-T therapy by Sorrento Therapeutics, anti-CD19/BCMA CAR-T cell therapy by Hrain Biotechnology, BCMA CAR-T therapy by Chineo Med (Beijing), BCMA TAC-T cell therapy by Triumvira Immunologies, BCMA-CAR T cell therapy by Shanghai Unicar-Therapy Biomed, BCMA/CD3 antibody by Regeneron, CAR-NK cell therapies by NantKwest, CC-93629, CMD-505, CTX-4419, CY AD- 211, HDP-101, HPN-217, P-BCMA-ALLO1, TNB-383B, bb-2121, AUTO-2, BCMA chimeric antigen receptor therapy by Pregene, BCMA-CAR T cells by Shanghai Bioray Laboratory, BCMA-CAR-T cells by CARsgen Therapeutics, CAR-T/TCR-T cell immunotherapy by Shenzhen BinDeBio, ET-140, P-BCMA-101, REGN-5458, AMG-701, anti BCMA CAR-T cell therapy by Cellular Biomedicine Group, bb-21217, BL836909, CC-93269, Descartes-08, IM-21, JNJ-64007957, MEDL2228 or PF-06863135.
[0100] In some embodiments, the BCMA x CD3 bispecific antibody can be, but is not limited to, elranatamab (also named PF-06863135), teneobio (also named TNB-383B), REGN5458, REGN5459, pavurutamab (also named AMG-701), BI 836909, CC-93269, WVT078 or teclistamab (also named JNJ-957 or JNJ-64007957). In some embodiments, the BCMA x CD3 bispecific antibody is elranatamab. In some embodiments, the BCMA x CD3 bispecific antibody is teneobio. In some embodiments, the BCMA x CD3 bispecific antibody is REGN5458. In some embodiments, the BCMA x CD3 bispecific antibody is REGN5459. In some embodiments, the BCMA x CD3 bispecific antibody is pavurutamab. In some embodiments, the BCMA x CD3 bispecific antibody is BI 836909. In some embodiments, the BCMA x CD3 bispecific antibody is CC-93269. In some embodiments, the BCMA x CD3 bispecific antibody is WVT078. In some embodiments, the BCMA x CD3 bispecific antibody is teclistamab.
[0101] In some embodiments, teclistamab comprises a first heavy chain (HC1), a first light chain (LC1), a second heavy chain (HC2), and a second light chain (LC2), wherein the HC1 is associated with LC 1 and the HC2 is associated with LC2, wherein HC 1 and LC 1 form a first antigen-binding site that immunospecifically binds to BCMA and wherein HC2 and LC2 form a second antigen-binding site that immunospecifically binds to CD3. In some embodiments, teclistamab comprises a HC1 of SEQ ID NO: 22, a LC1 of SEQ ID NO: 23, a HC2 of SEQ ID NO: 24, and a LC2 of SEQ ID NO: 25. In some embodiments, the BCMA arm and the CD3 arm of teclistamab form a functional bispecific antibody through an interaction of their respective Fc domains.
[0102] In some embodiments, the BCMA x CD3 bispecific antibody comprises any one of the BCMA binding domains described in Int. Pat. Publ. No. W02017/031104.
[0103] In some embodiments, the BCMA x CD3 bispecific antibody is an IgGl, an IgG2, an IgG3, or an IgG4 isotype.
[0104] In some embodiments, the BCMA x CD3 bispecific antibody is an IgGl isotype.
[0105] In some embodiments, the BCMA x CD3 bispecific antibody is an IgG2 isotype.
[0106] In some embodiments, the BCMA x CD3 bispecific antibody is an IgG3 isotype.
[0107] In some embodiments, the BCMA x CD3 bispecific antibody is an IgG4 isotype.
[0108] The BCMA x CD3 bispecific antibody can be of any allotype. It is expected that allotype has no influence on properties of the BCMA x CD3 bispecific antibodies, such as binding or Fc- mediated effector functions. Immunogenicity of therapeutic antibodies is associated with increased risk of infusion reactions and decreased duration of therapeutic response (Baert et al., (2003) N Engl J Med 348:602-08). The extent to which therapeutic antibodies induce an immune response in the host may be determined in part by the allotype of the antibody (Stickler et al., (2011) Genes and Immunity 12:213-21). Antibody allotype is related to amino acid sequence variations at specific locations in the constant region sequences of the antibody. Table 8 shows select IgGl, IgG2, and IgG4 allotypes.
TABLE 8: IgGl, IgG2 and IgG4 allotypes.
Figure imgf000033_0001
[0109] In some embodiments, the multispecific antibody comprises one or more Fc substitutions that reduces binding of the multispecific antibody to a Fey receptor (FcyR). Substitutions that reduce binding of the multispecific antibody to the FcyR reduces the Fc effector functions such as ADCC, ADCP and/or CDC of the multispecific antibody. The specific substitutions can be made in comparison to the wild-type IgGl of SEQ ID NO: 26 or the wild-type IgG4 of SEQ ID NO: 27.
[0110] In some embodiments, the one or more Fc substitutions is selected from the group consisting of F234A/L235A on IgG4, L234A/L235A on IgGl, V234A/G237A/ P238S/H268A/V309L/A330S/P331S on IgG2, F234A/L235A on IgG4, S228P/F234A/ L235A on IgG4, N297A on all Ig isotypes, V234A/G237A on IgG2, K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl, H268Q/V309L/A330S/P331S on IgG2, S267E/L328F on IgGl, L234F/L235E/D265A on IgGl, L234A/L235A/G237A/P238S/H268A/A330S/P331S on IgGl, S228P/F234A/L235A/G237A/P238S on IgG4 and S228P/F234A/L235A/G236- deleted/G237A/P238S on IgG4, wherein residue numbering is according to the EU index.
[0111] In some embodiments, the one or more Fc substitutions is F234A/L235A on IgG4.
[0112] In some embodiments, the one or more Fc substitutions is L234A/L235A on IgGl. [0113] In some embodiments, the one or more Fc substitutions is V234A/G237A/ P238S/H268A/V309L/A330S/P331S on IgG2.
[0114] In some embodiments, the one or more Fc substitutions is F234A/L235A on IgG4.
[0115] In some embodiments, the one or more Fc substitutions is S228P/F234A/ L235A on IgG4.
[0116] In some embodiments, the one or more Fc substitutions is N297A on all Ig isotypes.
[0117] In some embodiments, the one or more Fc substitutions is V234A/G237A on IgG2.
[0118] In some embodiments, the one or more Fc substitutions is K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl.
[0119] In some embodiments, the one or more Fc substitutions is H268Q/V309L/A330S/ P331S on IgG2.
[0120] In some embodiments, the one or more Fc substitutions is S267E/L328F on IgGl. In some embodiments, the one or more Fc substitutions is L234F/L235E/D265A on IgGl.
[0121] In some embodiments, the one or more Fc substitutions is L234A/L235A/G237A/ P238S/H268A/A330S/P331S on IgGl.
[0122] In some embodiments, the one or more Fc substitutions is S228P/F234A/L235A/ G237A/P238S on IgG4 and S228P/F234A/L235A/G236-deleted/G237A/P238S on IgG4.
[0123] In some embodiments, the bispecific antibody further comprises a S228P substitution.
[0124] In some embodiments, the bispecific antibody comprises one or more asymmetric substitutions in a first CH3 domain or in a second CH3 domain, or in both the first CH3 domain and the second CH3 domain.
[0125] In some embodiments, the one or more asymmetric substitutions is selected from the group consisting of F450L/K409R, wild-type/F409L_R409K, T366Y/F405A, T366W/F405W, F405W/Y407A, T394W/Y407T, T394S/Y407A, T366W/T394S, F405W/T394S and T366W/T366S_L368A_Y407V, L351 Y_F405 A_Y407V/T394W, T366I_K392M_T394W/F405A_Y407V, T366L_K392M_T394W/F405A_Y407V, L351Y_Y407A/T366A_K409F, L351Y_Y407A/T366V_K409F, Y407A/T366A_K409F and T350V_L351 Y_F405 A_Y407 V/T350V_T366L_K392L_T394W. [0126] In some embodiments, the one or more asymmetric substitutions is F450L/K409R. In some embodiments, the one or more asymmetric substitutions is wild-type/F409L_R409K. In some embodiments, the one or more asymmetric substitutions is T366Y/F405A. In some embodiments, the one or more asymmetric substitutions is T366W/F405W. In some embodiments, the one or more asymmetric substitutions is F405W/Y407A. In some embodiments, the one or more asymmetric substitutions is T394W/Y407T. In some embodiments, the one or more asymmetric substitutions is T394S/Y407A. In some embodiments, the one or more asymmetric substitutions is T366W/T394S. In some embodiments, the one or more asymmetric substitutions is F405W/T394S. In some embodiments, the one or more asymmetric substitutions is T366W/T366S_E368A_Y407V. In some embodiments, the one or more asymmetric substitutions is E351Y_F405A_Y407V/T394W. In some embodiments, the one or more asymmetric substitutions is T366I_K392M_T394W/F405A_Y407V. In some embodiments, the one or more asymmetric substitutions is T366E_K392M_T394W/F405A_Y407V. In some embodiments, the one or more asymmetric substitutions is E351Y_Y407A/T366A_K409F. In some embodiments, the one or more asymmetric substitutions is E351Y_Y407A/T366V_K409F. In some embodiments, the one or more asymmetric substitutions is Y407A/T366A_K409F. In some embodiments, the one or more asymmetric substitutions is
T350V_E351 Y_F405 A_Y407 V/T350V_T366E_K392E_T394W.
[0127] In some embodiments, the BCMA x CD3 bispecific antibody is an IgG4 isotype and comprises phenylalanine at position 405 and arginine at position 409 in a first heavy chain (HC1) and leucine at position 405 and lysine at position 409 in a second heavy chain (HC2), wherein residue numbering is according to the EU Index.
[0128] In some embodiments, the BCMA x CD3 bispecific antibody further comprises proline at position 228, alanine at position 234 and alanine at position 235 in both the HC1 and the HC2.
[0129] The present application describes various methods utilizing a BCMA x CD3 bispecific antibody. It can be any BCMA x CD3 bispecific antibody as provided herein, but it should be understood that the antibody in any of the methods provided for herein can be teclistamab. Anti-CD3 antibodies
[0130] CD38 is a multifunctional protein having function in receptor-mediated adhesion and signaling as well as mediating calcium mobilization via its ecto-enzymatic activity, catalyzing formation of cyclic ADP-ribose (cADPR) and AD PR. CD38 mediates cytokine secretion and activation and proliferation of lymphocytes (Funaro et al., J Immunol 145:2390-6, 1990;
Terhorst et al., Cell 771-80, 1981; Guse et al., Nature 398:70-3, 1999). CD38, via its NAD glycohydrolase activity, also regulates extracellular NAD+ levels, which have been implicated in modulating the regulatory T-cell compartment (Adriouch et al., Microbes infect 14: 1284-92, 2012; Chiarugi et al., Nature Reviews 12:741-52, 2012). In addition to signaling via Ca2+, CD38 signaling occurs via cross-talk with antigen-receptor complexes on T- and B -cells or other types of receptor complexes, e.g., major histocompatibility complex (MHC) molecules, involving CD38 in several cellular responses, but also in switching and secretion of IgGl.
[0131] Any suitable anti-CD38 antibody can be used in a method of the application.
[0132] In some embodiments, the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 28, the HCDR2 of SEQ ID NO: 29, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
[0133] The CDRs recited in above are of the Kabat numbering system. However, as provided for herein, the CDRs of the present disclosure may be provided by any appropriate numbering system, such as any of the Kabat, Chothia, IMGT or AbM numbering systems. Table 9 below provides exemplary CDRs utilizing the Kabat, Chothia, AbM, and IMGT numbering systems:
TABLE 9: Exemplary CDRs of the anti-CD38 antibody.
Figure imgf000036_0001
Figure imgf000037_0001
[0134] In some embodiments, the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 28, the HCDR2 of SEQ ID NO: 29, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
[0135] In some embodiments, the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 62, the HCDR2 of SEQ ID NO: 63, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
[0136] In some embodiments, the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 64, the HCDR2 of SEQ ID NO: 65, the HCDR3 of SEQ ID NO: 30, the LCDR1 of SEQ ID NO: 31, the LCDR2 of SEQ ID NO: 32 and the LCDR3 of SEQ ID NO: 33.
[0137] In some embodiments, the anti-CD38 antibody comprises the HCDR1 of SEQ ID NO: 66, the HCDR2 of SEQ ID NO: 67, the HCDR3 of SEQ ID NO: 68, the LCDR1 of SEQ ID NO: 69, an LCDR2 comprising the amino acid sequence of DA, and the LCDR3 of SEQ ID NO: 33.
[0138] In some embodiments, the anti-CD38 antibody comprises the VH of SEQ ID NO: 34 and the VL of SEQ ID NO: 35.
[0139] In some embodiments, the anti-CD38 antibody comprises the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37.
[0140] Other anti-CD38 antibodies used in the methods of the invention may be known antibodies, such as mAb003 described in U.S. Pat. No. 7,829,673. The VH and the VL of mAb003 may be expressed as IgGl/K; mAbO24 described in U.S. Pat. No. 7,829,673. The VH and the VL of mAbO24 may be expressed as IgGl/K; MOR-202 (MOR-03087) comprising described in US. Pat. No. 8,088,896. The VH and the VL of MOR-202 may be expressed as IgGl/K; or isatuximab; described in U.S. Pat. No. 8,153,765. The VH and the VL of isatuximab may be expressed as IgGl/K. In some embodiments, the anti-CD38 antibody comprises a) the VH of SEQ ID NO: 38 and the VL of SEQ ID NO: 39; b) the VH of SEQ ID NO: 40 and the VL of SEQ ID NO: 41 ; c) the VH of SEQ ID NO: 42 and the VL of SEQ ID NO: 43; or d) the VH of SEQ ID NO: 44 and the VL of SEQ ID NO: 45.
[0141] In some embodiments, the anti-CD38 antibody is DARZALEX® (daratumumab).
[0142] In some embodiments, daratumumab comprises the VH of SEQ ID NO: 34 and the VL of SEQ ID NO: 35.
[0143] In some embodiments, daratumumab comprises the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37.
[0144] In some embodiments, the anti-CD38 antibody is chimeric, humanized, or human.
[0145] In some embodiments, the anti-CD38 antibody is an IgGl, an IgG2, an IgG3, or an IgG4 isotype.
[0146] In some embodiments, the anti-CD38 antibody is an IgGl isotype.
[0147] In some embodiments, the anti-CD38 antibody is an antigen binding fragment. Exemplary antigen binding fragments are Fab, F(ab')2, Fd and Fv fragments.
[0148] The present application describes various methods utilizing an anti-CD38 antibody. It can be any anti-CD38 antibody as provided herein, but it should be understood that the antibody in any of the methods provided for herein can be daratumumab.
Immunomodulatory Drugs (IMiD)
[0149] Immunomodulatory drugs are known in the art and any appropriate immunomodulatory drug may be used in any of the embodiments provided herein. In some embodiments, the immunomodulatory drug is an immunomodulatory imide drug, such as but not limited to thalidomide, lenalidomide, pomalidomide, iberdomide, and apremilast. In some embodiments, the immunomodulatory drug is thalidomide. In some embodiments, the immunomodulatory drug is lenalidomide. In some embodiments, the immunomodulatory drug is pomalidomide. In some embodiments, the immunomodulatory drug is iberdomide. In some embodiments, the immunomodulatory drug is apremilast. [0150] The present application describes various methods utilizing an IMiD. It can be any IMiD as provided herein, but it should be understood that the IMiD in any of the methods provided for herein can be lenalidomide.
BCMA x CD3 bispecific antibody dosing
[0151] In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any amount administered for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 60 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 240 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 355 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 475 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 635 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 720 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 845 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1125 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1500 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 1685 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 2250 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 6000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
[0152] In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0153] In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the effective amount of the BCMA x CD3 bispecific antibody is any threshold. In some embodiments, the predetermined weight threshold is 50 kg, 55 kg, 60 kg, 65 kg, 70 kg, greater than 70 kg, or any value in-between.
[0154] In some embodiments, the pre-determined weight threshold is 50 kg, wherein those subjects who are < 50 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 50 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the pre- determined weight threshold is 55 kg, wherein those subjects who are < 55 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 55 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 60 kg, wherein those subjects who are < 60 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 60 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the predetermined weight threshold is 65 kg, wherein those subjects who are < 65 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 65 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 70 kg, wherein those subjects who are < 70 kg are in tier 1 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, and those subjects who are > 70 kg are in tier 2 and administered a therapeutically effective amount of the BCMA x CD3 bispecific antibody, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different.
[0155] In some embodiments, subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range inbetween administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0156] In some embodiments, subject is in tier 2 and the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg or any value or range inbetween administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of the BCMA x CD3 bispecific antibody is any value including or inbetween the values as provided for herein.
[0157] In some embodiments, the BCMA x CD3 bispecific antibody is any BCMA x CD3 bispecific antibody as provided for herein. In some embodiments, the BCMA x CD3 bispecific antibody is teclistamab. In some embodiments, teclistamab comprises an amino acid sequence as provided for herein. In some embodiments, the therapeutically effective amount of teclistamab is any amount administered for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of teclistamab is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 60 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 240 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 300 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 355 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 475 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 635 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 720 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 845 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 1125 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 1500 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 1685 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 2250 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 3000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 6000 pg/kg administered once every day, once a week, biweekly, triweekly, or once every four weeks.
[0158] In some embodiments, the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of teclistamab is any value including or in-between the values as provided for herein.
[0159] In some embodiments, the therapeutically effective amount of teclistamab is determined based on the weight of the subject. In some embodiments, the therapeutically effective amount of teclistamab is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered a therapeutically effective amount of teclistamab, and wherein if the subject is above the predetermined weight threshold then the subject is in tier 2 and is administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different. In some embodiments, the therapeutically effective amount of teclistamab for those subjects in tier 1 and the effective amount of teclistamab for those subjects in tier 2 are the same. In some embodiments, the effective amount of teclistamab for those subjects in tier 1 and the effective amount of teclistamab for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the effective amount of teclistamab is any threshold. In some embodiments, the pre-determined weight threshold is 50 kg, 55 kg, 60 kg, 65 kg, 70 kg, greater than 70 kg, or any value in-between. [0160] In some embodiments, the pre-determined weight threshold is 50 kg, wherein those subjects who are < 50 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 50 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 55 kg, wherein those subjects who are < 55 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 55 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 60 kg, wherein those subjects who are < 60 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 60 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 65 kg, wherein those subjects who are < 65 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 65 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different. In some embodiments, the pre-determined weight threshold is 70 kg, wherein those subjects who are < 70 kg are in tier 1 and administered a therapeutically effective amount of teclistamab, and those subjects who are > 70 kg are in tier 2 and administered a therapeutically effective amount of teclistamab, wherein the therapeutically effective amount of teclistamab for those subjects in tier 1 and the therapeutically effective amount of teclistamab for those subjects in tier 2 may be the same or different.
[0161] In some embodiments, subject is in tier 1 and the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 1 and the therapeutically effective amount of teclistamab is any value including or in-between the values as provided for herein.
[0162] In some embodiments, subject is in tier 2 and the therapeutically effective amount of teclistamab is from about 3 mg to about 600 mg or any value or range in-between administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 3 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 4 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 15 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 24 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 25 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 100 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 150 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 200 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 300 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is 450 mg administered once every day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the subject is in tier 2 and the effective amount of teclistamab is any value including or in-between the values as provided for herein.
[0163] The present application describes various methods utilizing a BCMA x CD3 bispecific antibody. It can be any BCMA x CD3 bispecific antibody as provided herein, but it should be understood that the antibody in any of the methods can be teclistamab.
CD38 antibody dosing
[0164] In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is any amount administered for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg, or any value or range in-between administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1400 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1500 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1600 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1700 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1900 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2000 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2100 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 2400 mg administered once a week, biweekly, triweekly, or once every four weeks.
[0165] In some embodiments, the anti-CD38 antibody is FDA-approved for clinical use. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks.
[0166] In some embodiments, the anti-CD38 antibody can be any anti-CD38 antibody as provided for herein. In some embodiments, the anti-CD38 antibody is daratumumab. In some embodiments, daratumumab comprises the amino acid sequences as provided for herein. In some embodiments, the therapeutically effective amount of daratumumab is any amount administered for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of daratumumab is from about 1200 mg to about 2400 mg, or any value or range in-between administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1400 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1500 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1600 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1700 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 1900 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2000 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2100 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2200 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2300 mg administered once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of daratumumab is 2400 mg administered once a week, biweekly, triweekly, or once every four weeks.
[0167] In some embodiments, the therapeutically effective amount of daratumumab is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of daratumumab is 1800 mg administered once a week, biweekly, triweekly, or once every four weeks.
[0168] The present application describes various methods utilizing an anti-CD38 antibody. It can be any anti-CD38 antibody as provided herein, but it should be understood that the antibody in any of the methods can be daratumumab. IMiD dosing
[0169] In some embodiments, the therapeutically effective amount of the IMiD is any amount administered for a time sufficient to treat the cancer. In some embodiments, the effective amount of the IMiD is from about 15 mg to about 50 mg, or any value or range in-between administered once a day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of the IMiD is 25 mg administered daily. In some embodiments, the IMiD is FDA approved for clinical use. In some embodiments, the therapeutically effective amount of the IMiD is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of the IMiD is 25 mg.
[0170] In some embodiments, the IMiD is any IMiD as provided for herein. In some embodiments, the IMiD is lenalidomide. In some embodiments, the therapeutically effective amount of lenalidomide is any amount administered for a time sufficient to treat the cancer. In some embodiments, the effective amount of lenalidomide is from about 15 mg to about 50 mg, or any value or range in-between administered once a day, once a week, biweekly, triweekly, or once every four weeks. In some embodiments, the therapeutically effective amount of lenalidomide is 25 mg administered daily. In some embodiments, the therapeutically effective amount of lenalidomide is based on the standard doses used in clinical practices for the treatment of cancer. In some embodiments, the therapeutically effective amount of lenalidomide is 25 mg.
Routes of administration
[0171] The BCMA x CD3 bispecific antibody, anti-CD38 antibody, and IMiD of the methods disclosed herein may be administered to the subject via any appropriate method of administration. Exemplary methods of administration include, but are not limited to transarterial, subcutaneous, intradermal, intratumoral, intraanodal, intramedullary, intramuscular, intravenous, intraperitoneal, intranasal, or intraosseous. In some embodiments, the BCMA x CD3 bispecific antibody is administered intravenously or subcutaneously. In some embodiments, the BCMA x CD3 bispecific antibody is administered intravenously (i.v.). In some embodiments, the BCMA x CD3 bispecific antibody is administered subcutaneously (s.c.). In some embodiments, the anti-CD38 bispecific antibody is administered intravenously or subcutaneously. In some embodiments, the anti-CD38 bispecific antibody is administered intravenously (i.v.). In some embodiments, the anti-CD38 bispecific antibody is administered subcutaneously (s.c.). In some embodiments, the IMiD bispecific antibody is administered intravenously or orally. In some embodiments, the IMiD bispecific antibody is administered intravenously (i.v.). In some embodiments, the IMiD bispecific antibody is administered orally.
[0172] In some embodiments, the method comprises intravenously administering an therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, intravenously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises intravenously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises intravenously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and intravenously administering a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprises subcutaneously administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, subcutaneously administering a therapeutically effective amount of an anti-CD38 antibody, and orally administering a therapeutically effective amount of an immunomodulatory drug (IMiD) (oral) to the subject for a time sufficient to treat the cancer.
Dose Timing
[0173] The components of the methods provided for herein may be provided at any dose or frequency required to effectively treat the cancer. In some embodiments, the therapeutically effective amount of the components of the methods provided for herein may be provided at any frequency required to effectively treat the cancer.
[0174] In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered weekly. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered biweekly. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once every three weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once every four weeks. In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered once a month. The frequency of administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be altered as necessary to effectively treat the cancer. For example, the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
[0175] In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered daily. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered weekly. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered biweekly. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered once every three weeks. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered once every four weeks. In some embodiments, the therapeutically effective amount of the anti- CD38 antibody is administered once a month. The frequency of administration of the therapeutically effective amount of the anti-CD38 antibody may be altered as necessary to effectively treat the cancer. For example, the therapeutically effective amount of the anti-CD38 antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the anti-CD38 antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
[0176] In some embodiments, the therapeutically effective amount of the IMiD is administered daily. In some embodiments, the therapeutically effective amount of the IMiD is administered weekly. In some embodiments, the therapeutically effective amount of the IMiD is administered biweekly. In some embodiments, the therapeutically effective amount of the IMiD is administered once every three weeks. In some embodiments, the therapeutically effective amount of the IMiD is administered once every four weeks. In some embodiments, the therapeutically effective amount of the IMiD is administered once a month. The frequency of administration of the therapeutically effective amount of the IMiD may be altered as necessary to effectively treat the cancer. For example, the therapeutically effective amount of the IMiD may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the therapeutically effective amount of the IMiD may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
[0177] In some embodiments, the therapeutically effective amount and frequency of a component of the method is held constant for a recurring period of time, or a cycle. In some embodiments, the duration of a treatment cycle may be for any period of time. In some embodiments, a treatment cycle may last for 7, 8, 9, 10, 11, 12, 13, 14, 21, 28, 56, 112, 224, 365, more than 365 days, or any number of days in-between. In some embodiments, the treatment cycle lasts for 28 days. It is to be understood that the length of time of a treatment cycle is not meant to limit the duration of the treatment in any way. The treatment may continue for any number of treatment cycles necessary to treat the cancer. In some embodiments, the components of the methods provided for herein may be administered to the subject for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more than 20 treatment cycles, or the components of the methods provided for herein are administered to the subject until the cancer is treated. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for two treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for three treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for four treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for five treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for six treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for seven treatment cycles. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for more than seven treatment cycles. Further, it is to be understood, as provided for herein, that the therapeutically effective amount or frequency of the components of the methods provided for herein may be altered at the conclusion of each treatment cycle. Accordingly, in some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for two treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for three treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti- CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for four treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for five treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for six treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for seven treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti- CD38 antibody, and the IMiD may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for more than seven treatment cycles, wherein the therapeutically effective amount or frequency of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD may or may not be changed between each treatment cycle.
[0178] In some embodiments, the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered weekly for a first and second treatment cycles, biweekly for a third, fourth, fifth, and sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles. In some embodiments, the therapeutically effective amount of the anti-CD38 antibody is administered weekly for a first and second treatment cycles, biweekly for a third, fourth, fifth, and sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles. In some embodiments, the therapeutically effective amount of the IMiD is administered daily for days 1-21 of a 28-day treatment cycle. In some embodiments, the therapeutically effective amount of the IMiD is not administered in treatment cycle one. In some embodiments, the therapeutically effective amount of the IMiD is administered starting at treatment cycle two.
[0179] The duration of the treatment of the method may continue for any amount of time necessary to treat the cancer. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, 48, 52, 104, 156, 208, 260 or more weeks, or any number of weeks in-between. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 260 weeks. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or more months, or any number of months in-between. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 60 months. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years, or any number of years inbetween. In some embodiments, the subject is treated with therapeutically effective amounts of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD for more than 10 years.
Loading and Treatment Doses
[0180] In some embodiments, components of the methods provided for herein may be provided as a loading dose, a treatment dose, or a combination thereof. In some embodiments, components of the methods provided for herein are provided at a treatment dose. In some embodiments, components of the methods provided for herein are provided at a loading dose followed by a treatment dose. In some embodiments, the loading dose comprises one or more step-up doses provided over a period of time. In some embodiments, one step-up dose is provided. In some embodiments, two step-up doses are provided. In some embodiments, three step-up doses are provided. In some embodiments, more than three step-up doses are provided. In some embodiments, any number of step-up doses may be provided as necessary in order to provide the subject the effective amount of the components necessary to treat the cancer.
[0181] In some embodiments, the BCMA x CD3 bispecific antibody may be administered at a loading dose followed by a treatment dose. In some embodiments, the loading dose comprises one or more step-up doses as provided for herein. In some embodiments, the BCMA x CD3 bispecific antibody is administered as one step-up dose followed by a treatment dose. In some embodiments, the BCMA x CD3 bispecific antibody is administered as two step-up doses followed by a treatment dose. In some embodiments, the BCMA x CD3 bispecific antibody is administered as three step-up doses followed by a treatment dose. In some embodiments, the BCMA x CD3 bispecific antibody is administered as more than three step-up doses followed by a treatment dose.
[0182] In some embodiments, a first step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg, or any value or range in-between. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 300 Pg/kg-
[0183] In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg. [0184] In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an first step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an first step-up dose of the BCMA x CD3 bispecific antibody, wherein the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the first step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
[0185] In some embodiments, subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0186] In some embodiments, subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0187] In some embodiments, second step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 300 pg/kg.
[0188] In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the first step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0189] In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an second step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an second step-up dose of the BCMA x CD3 bispecific antibody, wherein the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the second step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
[0190] In some embodiments, subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0191] In some embodiments, subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the second step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0192] In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 300 pg/kg.
[0193] In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0194] In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody is determined based on a predetermined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an third or greater step-up dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an third or greater step-up dose of the BCMA x CD3 bispecific antibody, wherein the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the pre-determined weight threshold to determine the third or greater step-up dose of the BCMA x CD3 bispecific antibody is as provided for herein.
[0195] In some embodiments, subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range inbetween. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 1 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0196] In some embodiments, subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range inbetween. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the subject is in tier 2 and the third or greater step-up dose of the BCMA x CD3 bispecific antibody is 25 mg.
[0197] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose A and comprise a dose from about 60 pg/kg to about 150 pg/kg, or any value or range in-between. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 70 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 80 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 90 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 100 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 125 pg/kg. In some embodiments, the Step-up dose A of the BCMA x CD3 bispecific antibody is 150 pg/kg.
[0198] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose B and comprise a dose from about 150 pg/kg to about 250 or any value or range in-between. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 150 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 160 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 170 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 180 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 190 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 200 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 210 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 220 pg/kg. In some embodiments, the Step- up dose B of the BCMA x CD3 bispecific antibody is 230 pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 240pg/kg. In some embodiments, the Step-up dose B of the BCMA x CD3 bispecific antibody is 250pg/kg.
[0199] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose C and comprise a dose from about 5 pg/kg to about 60 pg/kg, or any value or range in-between. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 5 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 10 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 15 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 20 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 25 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 30 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody IS 40 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody is 50 pg/kg. In some embodiments, the Step-up dose C of the BCMA x CD3 bispecific antibody is 60 pg/kg.
[0200] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose D and comprise a dose from about 250 pg/kg to about 600 pg/kg, or any value or range in-between. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 250 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 300 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 350 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 400 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 450 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 500 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 550 pg/kg. In some embodiments, the Step-up dose D of the BCMA x CD3 bispecific antibody is 600 pg/kg. [0201] In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody of the methods provided for herein may comprise one or more of any dose or range selected from Step- up Dose A, B, C, D, or any combination thereof. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up doses as provided for in Table 10 below:
TABLE 10
Figure imgf000066_0001
Figure imgf000067_0001
[0202] In some embodiments, a step-up doses of the BCMA x CD3 bispecific antibody may be designated as Step-up dose E and comprise a dose from about 3 mg to about 12 mg, or any value or range in-between. In some embodiments, the Step-up dose E of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the Step-up dose E of the BCMA x CD3 bispecific antibody is 4 mg.
[0203] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose F and comprise a dose from about 12 mg to about 20 mg, or any value or range in-between. In some embodiments, the Step-up dose F of the BCMA x CD3 bispecific antibody is 12 mg. In some embodiments, the Step-up dose F of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the Step-up dose F of the BCMA x CD3 bispecific antibody is 20 mg.
[0204] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose G and comprise a dose from about 20 mg to about 30 mg, or any value or range in-between. In some embodiments, the Step-up dose G of the BCMA x CD3 bispecific antibody is 20 mg. In some embodiments, the Step-up dose G of the BCMA x CD3 bispecific antibody is 24 mg. In some embodiments, the Step-up dose G of the BCMA x CD3 bispecific antibody is 25 mg. In some embodiments, the Step-up dose G of the BCMA x CD3 bispecific antibody is 30 mg.
[0205] In some embodiments, a step-up dose of the BCMA x CD3 bispecific antibody may be designated as Step-up dose H and comprise a dose from about 30 mg to about 50 mg, or any value or range in-between. In some embodiments, the Step-up dose H of the BCMA x CD3 bispecific antibody is 30 mg. In some embodiments, the Step-up dose H of the BCMA x CD3 bispecific antibody is 40 mg. In some embodiments, the Step-up dose H of the BCMA x CD3 bispecific antibody is 50 mg.
[0206] In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody of the methods provided for herein may comprise one or more of any dose or range selected from Step- up Dose E, F, G, or H, or any combination thereof. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up doses as provided for in Table 11 below:
TABLE 11
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
[0207] It is to be understood that in embodiments wherein the BCMA x CD3 bispecific antibody is administered at more than one step-up dose, the current step-up dose and subsequent step-up does(s) may comprise the same or different doses and may comprises the same or different frequencies. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise the same dose and same frequency. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise the same dose and different frequencies. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise different doses and the same frequency. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise different doses and different frequencies.
[0208] In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered at a frequency as provided for herein. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered every other day. In some embodiments, the loading dose of the BCMA x CD3 bispecific antibody is administered weekly.
[0209] As provided for herein, the loading dose of the BCMA x CD3 bispecific antibody may comprise one or more step-up dose(s). Accordingly, in some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered at a frequency as provided for herein. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered daily. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered every other day. In some embodiments, the one or more step-up dose(s) of the BCMA x CD3 bispecific antibody are administered weekly.
[0210] In some embodiments, the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 10. In some embodiments, the one or more step-up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered daily. In some embodiments, the one or more step- up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered every other day. In some embodiments, the one or more step-up dose(s) selected from step-up dose A, B, C, or D as provided for herein are administered weekly. When administered in combination, each step-up dose A, B, C, or D may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein. Accordingly, in some embodiments, a first step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group A, B, C, or D as the first step-up dose and wherein the first and second time periods may be the same or different. In embodiments comprising a third step-up dose, a third step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a third time period, wherein the first, second, and third step-up doses are not of the same group A, B, C, or D and wherein the first, second, and third time periods may be the same or different. In embodiments comprising a fourth step-up dose, a fourth step-up dose selected from A, B, C, or D as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a fourth time period, wherein the first, second, third, and fourth step-up doses are all of a unique group A, B, C, or D and wherein the first, second, third, and fourth time periods may be the same or different.
[0211] In some embodiments, the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 11. In some embodiments, the one or more step-up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered daily. In some embodiments, the one or more step- up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered every other day. In some embodiments, the one or more step-up dose(s) selected from step-up dose E, F, G, or H as provided for herein are administered weekly. When administered in combination, each step-up dose E, F, G, or H may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein. Accordingly, in some embodiments, a first step-up dose selected from E,
F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group E, F,
G, or H as the first step-up dose and wherein the first and second time periods may be the same or different. In embodiments comprising a third step-up dose, a third step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly month for a third time period, wherein the first, second, and third step-up doses are not of the same group E, F, G, or H and wherein the first, second, and third time periods may be the same or different. In embodiments comprising a fourth step-up dose, a fourth step-up dose selected from E, F, G, or H as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a fourth time period, wherein the first, second, third, and fourth step-up doses are not of the same group E, F, G, or H and wherein the first, second, third, and fourth time periods may be the same or different.
[0212] In some embodiments, treatment dose of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg or any value or range in-between. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 60 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 240 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 300 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 355 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 475 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 635 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 720 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 845 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1125 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1500 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 1685 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 2250 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 3000 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 6000 pg/kg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0213] In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0214] In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is determined based on the weight of the subject. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is determined based on a pre-determined weight threshold, wherein if the subject is at or below the pre-determined weight threshold then the subject is in tier 1 and administered an treatment dose of the BCMA x CD3 bispecific antibody, and wherein if the subject is above the pre-determined weight threshold then the subject is in tier 2 and is administered an treatment dose of the BCMA x CD3 bispecific antibody, wherein the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 may be the same or different In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are the same. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody for those subjects in tier 2 are different. In some embodiments, the predetermined weight threshold to determine the treatment dose of the BCMA x CD3 bispecific antibody is as provided for herein.
[0215] In some embodiments, subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the subject is in tier 1 and the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0216] In some embodiments, subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg, or any value or range in-between. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 3 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 4 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 15 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 25 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 100 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 150 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 200 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 300 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is 450 mg. In some embodiments, the subject is in tier 2 and the treatment dose of the BCMA x CD3 bispecific antibody is any value including or in-between the values as provided for herein.
[0217] In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered daily. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered weekly. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered biweekly. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once every three weeks. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once every four weeks. In some embodiments, the treatment dose of the BCMA x CD3 bispecific antibody is administered once a month. The frequency of administration of the treatment dose of the BCMA x CD3 bispecific antibody may be altered as necessary to treat the cancer. For example, the treatment dose of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the treatment dose of the BCMA x CD3 bispecific antibody may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
Co-treatments and pre-treatments
[0218] In some embodiments, the method of treating cancer comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen, a cotreatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer. In some embodiments, the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti- CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen. In some embodiments, the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a cotreatment regimen. In some embodiments, the method comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) further comprises administering a pre-treatment regimen and a co-treatment regimen. It is to be understood that any of the methods of the present disclosure may further comprise the administration of a pre-treatment regimen, a co-treatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer. Accordingly, in some embodiments, a method as provided for herein further comprises administering a pre-treatment regimen, a co-treatment regimen, or a combination thereof to the subject for a time sufficient to treat the cancer. In some embodiments, a method as provided for herein further comprises administering a pre-treatment regimen for a time sufficient to treat the cancer. In some embodiments, a method as provided for herein further comprises administering a co-treatment regimen for a time sufficient to treat the cancer. In some embodiments, a method as provided for herein further comprises administering a pre-treatment regimen and a co-treatment regimen to the subject for a time sufficient to treat the cancer.
[0219] It is to be understood that as used herein “pre-treatment regimen” can refer to a regimen of pharmaceutical compounds, compositions, or formulas that are administered prior to the start of the administration of a therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD). “Pre-treatment regimen” can also refer to refer to a regimen of pharmaceutical compounds, compositions, or formulas that are administered during the course of administration of a therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD), but wherein the pre-treatment regimen is administered prior to administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti- CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD) should dosing occur on the same day. Thus, it is to be understood that, as used herein, “pretreatment regimen” dose not exclude the components of the pre-treatment regimen from be administered during the course of administration of the therapeutically effective amount of the BCMA x CD3 bispecific antibody, a therapeutically effective amount of the anti-CD38 antibody, and a therapeutically effective amount of the immunomodulatory drug (IMiD).
[0220] In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, a therapeutically effective amount of an antipyretic, or any combination thereof. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of an antipyretic. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, and a therapeutically effective amount of an antihistamine. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid and a therapeutically effective amount of an antipyretic. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine and a therapeutically effective amount of an antipyretic. In some embodiments, the pre-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, and a therapeutically effective amount of an antipyretic.
[0221] In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, a therapeutically effective amount of an antipyretic, or any combination thereof. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of an antipyretic. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, and a therapeutically effective amount of an antihistamine. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid and a therapeutically effective amount of an antipyretic. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of an antihistamine and a therapeutically effective amount of an antipyretic. In some embodiments, the co-treatment regimen comprises administration of a therapeutically effective amount of a glucocorticoid, a therapeutically effective amount of an antihistamine, and a therapeutically effective amount of an antipyretic.
Glucocorticoid dosing
[0222] In some embodiments, the therapeutically effective amount of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of the glucocorticoid is from about 8 mg to about 50 mg or any value or range in-between. In some embodiments, the therapeutically effective amount of the glucocorticoid is 8 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 10 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 12 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 14 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid 16 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid 18 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 20 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 25 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 30 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 35 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 40 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 45 mg. In some embodiments, the therapeutically effective amount of the glucocorticoid is 50 mg. Antihistamine dosing
[0223] In some embodiments, the therapeutically effective amount of the antihistamine is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of the antihistamine is from about 25 mg to about 50 mg, or any value or range in-between, In some embodiments, the therapeutically effective amount of the antihistamine is 25 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 30 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 35 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 40 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 45 mg. In some embodiments, the therapeutically effective amount of the antihistamine is 50 mg.
Antipyretic dosing
[0224] In some embodiments, the therapeutically effective amount of the antipyretic is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the therapeutically effective amount of the antipyretic is from about 500 mg to about 1000 mg, or any value or range in-between. In some embodiments, the therapeutically effective amount of the antipyretic is 500 mg.
Figure imgf000079_0001
some embodiments, the therapeutically effective amount of the antipyretic is 550 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 600 mg.
Figure imgf000079_0002
some embodiments, the therapeutically effective amount of the antipyretic is 650 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 700 mg.
Figure imgf000079_0003
some embodiments, the therapeutically effective amount of the antipyretic is 750 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 800 mg.
Figure imgf000079_0004
some embodiments, the therapeutically effective amount of the antipyretic is 850 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 900 mg.
Figure imgf000079_0005
some embodiments, the therapeutically effective amount of the antipyretic is 950 mg. In some embodiments, the therapeutically effective amount of the antipyretic is 1000 mg. Routes of administration for pre-treatment and co-treatment regimens
[0225] The glucocorticoid, antihistamine, and antipyretic of the methods disclosed herein may be administered to the subject via any appropriate method of administration. Exemplary methods of administration include, but are not limited to transarterial, subcutaneous, intradermal, intratumoral, intraanodal, intramedullary, intramuscular, intravenous, intraperitoneal, intranasal, or intraosseous. In some embodiments, the glucocorticoid is administered orally or intravenously (i.v.). In some embodiments, the glucocorticoid is administered orally. In some embodiments, the glucocorticoid is administered intravenously (i.v.). In some embodiments, the antihistamine is administered orally or intravenously (i.v.). In some embodiments, the antihistamine is administered orally. In some embodiments, the antihistamine is administered intravenously (i.v.). In some embodiments, the antipyretic is administered orally or intravenously (i.v.). In some embodiments, the antipyretic is administered orally. In some embodiments, the antipyretic is administered intravenously (i.v.).
Frequency of administration for pre-treatment and co-treatment regimens
[0226] The components of the pre-treatment and co-treatment regimens of the methods provided for herein may be provided at any dose or frequency. In some embodiments, the therapeutically effective amount of the pre-treatment and co-treatment regimens may be provided at any frequency.
[0227] In some embodiments, the glucocorticoid is administered daily. In some embodiments, effective amount of the glucocorticoid is administered every other day. In some embodiments, effective amount of the glucocorticoid is administered weekly. In some embodiments, effective amount of the glucocorticoid is administered biweekly. In some embodiments, effective amount of the glucocorticoid is administered once every three weeks. In some embodiments, effective amount of the glucocorticoid is administered once every four weeks. In some embodiments, effective amount of the glucocorticoid is administered once a month. The frequency of administration of the effective amount of the glucocorticoid may be altered as necessary to achieve a desired effect. For example, the effective amount of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time.
[0228] In some embodiments, the antihistamine is administered daily. In some embodiments, effective amount of the antihistamine is administered every other day. In some embodiments, effective amount of the antihistamine is administered weekly. In some embodiments, effective amount of the antihistamine is administered biweekly. In some embodiments, effective amount of the antihistamine is administered once every three weeks. In some embodiments, effective amount of the antihistamine is administered once every four weeks. In some embodiments, effective amount of the antihistamine is administered once a month. The frequency of administration of the effective amount of the antihistamine may be altered as necessary to achieve a desired effect. For example, the effective amount of the antihistamine may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the antihistamine may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time.
[0229] In some embodiments, the antipyretic is administered daily. In some embodiments, effective amount of the antipyretic is administered every other day. In some embodiments, effective amount of the antipyretic is administered weekly. In some embodiments, effective amount of the antipyretic is administered biweekly. In some embodiments, effective amount of the antipyretic is administered once every three weeks. In some embodiments, effective amount of the antipyretic is administered once every four weeks. In some embodiments, effective amount of the antipyretic is administered once a month. The frequency of administration of the effective amount of the antipyretic may be altered as necessary to achieve a desired effect. For example, the effective amount of the antipyretic may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a certain period of time, after which time the effective amount of the antipyretic may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the recited frequencies for a next period of time. [0230] In some embodiments, the therapeutically effective amount and frequency of a component of the method is held constant for a recurring period of time, or a cycle. In some embodiments, the duration of a treatment cycle may be for any period of time. In some embodiments, a treatment cycle may last for 7, 8, 9, 10, 11, 12, 13, 14, 21, 28, 56, 112, 224, 365, more than 365 days, or any number of days in-between. In some embodiments, the treatment cycle lasts for 28 days. It is to be understood that the length of time of a treatment cycle is not meant to limit the duration of the treatment in any way. The treatment may continue for any number of treatment cycles necessary to treat the cancer. In some embodiments, the components of the methods provided for herein may be administered to the subject for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more than 20 treatment cycles, or the components of the methods provided for herein are administered to the subject until the cancer is treated. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for two treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for three treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for four treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for five treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for six treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for seven treatment cycles. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for more than seven treatment cycles.
[0231] It is to be understood, that any of the components herein may or may not be administered in any of the treatment cycles described herein. For example, in some embodiments, the therapeutically effective amount of the glucocorticoid may be administered for a first number of treatment cycles as provided for herein, the therapeutically effective amount of the antihistamine may be administered for a second number of treatment cycles, and the therapeutically effective amount of the antipyretic may be administered for a third number of treatment cycles, wherein the first, second and third number of treatment cycles may be the same or different.
[0232] Further, it is to be understood, as provided for herein, that the effective amount or frequency of the components of the methods provided for herein may be altered at the conclusion of each treatment cycle. Accordingly, in some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for one treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for two treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for three treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for four treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for five treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for six treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for seven treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle. In some embodiments, the therapeutically effective amounts of the glucocorticoid, the antihistamine, and the antipyretic are administered to the subject for more than seven treatment cycles, wherein the therapeutically effective amount or frequency of the glucocorticoid, the antihistamine, and the antipyretic may or may not be changed between each treatment cycle.
[0233] In some embodiments, the therapeutically effective amount of the glucocorticoid is administered on days 1, 2, 4, and 8 of a first treatment cycle, and then administered weekly for a second, third, and fourth treatment cycle. In some embodiments, the therapeutically effective amount of the antihistamine is administered weekly for a first and second treatment cycles, and then administered biweekly for a third, fourth, fifth, and sixth treatment cycle. In some embodiments, the therapeutically effective amount of the antipyretic is administered weekly for a first and second treatment cycles, and then administered biweekly for a third, fourth, fifth, and sixth treatment cycle.
[0234] In some embodiments, the therapeutically effective amount of the glucocorticoid is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered for a first, second, third, and fourth treatment cycles. In some embodiments, the therapeutically effective amount of the glucocorticoid is further administered after an administration relation reaction to the anti-CD38 antibody. In some embodiments, the therapeutically effective amount of the glucocorticoid is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody. In some embodiments, the therapeutically effective amount of the antihistamine is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered. In some embodiments, the therapeutically effective amount of the antihistamine is further administered after an administration relation reaction to the anti- CD38 antibody. In some embodiments, the therapeutically effective amount of the antihistamine is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody. In some embodiments, the therapeutically effective amount of the antipyretic is administered on all days the anti-CD38 antibody and/or the BCMAxCD3 bispecific antibody is administered. In some embodiments, the therapeutically effective amount of the antipyretic is further administered after an administration relation reaction to the anti-CD38 antibody. In some embodiments, the therapeutically effective amount of the antipyretic is further administered after an administration relation reaction to or a cytokine release syndrome caused by the BCMAxCD3 bispecific antibody.
[0235] The duration of the treatment of the method may continue for any amount of time necessary to treat the cancer. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, 48, 52, 104, 156, 208, 260 or more weeks, or any number of weeks inbetween. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 260 weeks. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or more months, or any number of months in-between. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 60 months. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years, or any number of years in-between. In some embodiments, the subject is treated with effective amounts of the glucocorticoid, the antihistamine, and the antipyretic for more than 10 years.
Loading and Treatment doses
[0236] In some embodiments, components of the method provided for herein may be provided as a loading dose, a treatment dose, or a combination thereof. In some embodiments, components of the methods provided for herein are provided at a treatment dose. In some embodiments, components of the methods provided for herein are provided at a loading dose followed by a treatment dose. In some embodiments, the loading dose comprises one or more step-up doses provided over a period of time. In some embodiments, one step-up dose is provided. In some embodiments, two step-up doses are provided. In some embodiments, three step-up doses are provided. In some embodiments, four step-up doses are provided. In some embodiments, more than four step-up doses are provided.
[0237] In some embodiments, the glucocorticoid may be administered at a loading dose followed by a treatment dose. In some embodiments, the loading dose comprises one or more step-up doses as provided for herein. In some embodiments, the glucocorticoid is administered as one step-up dose followed by a treatment dose. In some embodiments, the glucocorticoid is administered as two step-up doses followed by a treatment dose. In some embodiments, the glucocorticoid is administered as three step-up doses followed by a treatment dose. In some embodiments, the glucocorticoid is administered as four step-up doses followed by a treatment dose.
[0238] In some embodiments, the first step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the first step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the first step-up dose of the glucocorticoid is 8 mg. In some embodiments, the first step-up dose of the glucocorticoid is 10 mg. In some embodiments, the first step-up dose of the glucocorticoid is 12 mg. In some embodiments, the first step-up dose of the glucocorticoid is 14 mg. In some embodiments, the first step-up dose of the glucocorticoid is 16 mg. In some embodiments, the first step-up dose of the glucocorticoid is 18 mg. In some embodiments, the first step-up dose of the glucocorticoid is 20 mg. In some embodiments, the first step-up dose of the glucocorticoid is 25 mg. In some embodiments, the first step-up dose of the glucocorticoid is 30 mg. In some embodiments, the first step-up dose of the glucocorticoid is 35 mg. In some embodiments, the first step-up dose of the glucocorticoid is 40 mg. In some embodiments, the first step-up dose of the glucocorticoid is 45 mg. In some embodiments, the first step-up dose of the glucocorticoid is 50 mg.
[0239] In some embodiments, the second step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the second step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the second step-up dose of the glucocorticoid is 8 mg. In some embodiments, the second step-up dose of the glucocorticoid is 10 mg. In some embodiments, the second step-up dose of the glucocorticoid is 12 mg. In some embodiments, the second step-up dose of the glucocorticoid is 14 mg. In some embodiments, the second step-up dose of the glucocorticoid is 16 mg. In some embodiments, the second step-up dose of the glucocorticoid is 18 mg. In some embodiments, the second step-up dose of the glucocorticoid is 20 mg. In some embodiments, the second step-up dose of the glucocorticoid is
25 mg. In some embodiments, the second step-up dose of the glucocorticoid is 30 mg. In some embodiments, the second step-up dose of the glucocorticoid is 35 mg. In some embodiments, the second step-up dose of the glucocorticoid is 40 mg. In some embodiments, the second step-up dose of the glucocorticoid is 45 mg. In some embodiments, the second step-up dose of the glucocorticoid is 50 mg.
[0240] In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is any amount administered in combination with the treatment regimen for a time sufficient to treat the cancer. In some embodiments, the third, fourth or greater step-up dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 8 r ig. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 10 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 12 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 14 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 16 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 18 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 20 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 25 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 30 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 35 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 40 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 45 mg. In some embodiments, the third, fourth, or greater step-up dose of the glucocorticoid is 50 mg.
[0241] In some embodiments, a step-up dose of the glucocorticoid may be designated as Step-up dose J and comprise a dose from about, about 8 mg to about 12 mg, or any value or range inbetween. In some embodiments, the Step-up dose J of the glucocorticoid is 8 mg. In some embodiments, the Step-up dose J of the glucocorticoid is 9 mg. In some embodiments, the Step- up dose J of the glucocorticoid is 10 mg. In some embodiments, the Step-up dose J of the glucocorticoid is 11 mg. In some embodiments, the Step-up dose J of the glucocorticoid is 12 mg.
[0242] In some embodiments, a step-up dose of the glucocorticoid may be designated as Step-up dose K and comprise a dose from about 12 mg to about 20 mg, or any value or range in-between. In some embodiments, the Step-up dose K of the glucocorticoid is 12 mg. In some embodiments, the Step-up dose K of the glucocorticoid is 14 mg. In some embodiments, the Step-up dose K of the glucocorticoid is 16 mg. In some embodiments, the Step-up dose K of the glucocorticoid is 18 mg. In some embodiments, the Step-up dose K of the glucocorticoid is 20 mg.
[0243] In some embodiments, a step-up dose of the glucocorticoid may be designated as Step-up dose L and comprise a dose from about 20 mg to about 50 mg, or any value or range in-between. In some embodiments, the Step-up dose L of the glucocorticoid is 20 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 25 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 30 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 35 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 40 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 45 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 50 mg. In some embodiments, the Step-up dose L of the glucocorticoid is 60 mg. In some
[0244] In some embodiments, the loading dose of the glucocorticoid of the methods provided for herein may comprise one or more of any dose or range selected from Step-up Dose J, K, L, or any combination thereof. In some embodiments, the loading dose of the glucocorticoid may comprise one or more step-up doses as provided for in Table 12 below:
TABLE 12
Figure imgf000088_0001
Figure imgf000089_0001
[0245] It is to be understood that in embodiments wherein the glucocorticoid is administered at more than one step-up dose, the current step-up dose and subsequent step-up does(s) may comprise the same or different doses and may comprises the same or different frequencies. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise the same dose and same frequency. In some embodiments, the current step-up dose and subsequent step- up dose(s) comprise the same dose and different frequencies. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise different doses and the same frequency. In some embodiments, the current step-up dose and subsequent step-up dose(s) comprise different doses and different frequencies.
[0246] In some embodiments, the loading dose of the glucocorticoid is administered at a frequency as provided for herein. In some embodiments, the loading dose of the glucocorticoid is administered daily. In some embodiments, the loading dose of the glucocorticoid is administered every other day. In some embodiments, the loading dose of the glucocorticoid is administered weekly.
[0247] As provided for herein, the loading dose of the glucocorticoid may comprise one or more step-up dose(s). Accordingly, in some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered at a frequency as provided for herein. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered daily. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered every other day. In some embodiments, the one or more step-up dose(s) of the glucocorticoid are administered weekly.
[0248] In some embodiments, the loading dose comprises one or more step-up doses, wherein the one or more step-up dose(s) are administered at a dose and combination as outlined in Table 12. In some embodiments, the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered daily. In some embodiments, the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered every other day. In some embodiments, the one or more step-up dose(s) selected from step-up dose J, K, or L as provided for herein are administered weekly. When administered in combination, each step-up dose J, K, or L may be administered for any frequency as provided for herein prior to administration of the second step-up dose which may be administered for any frequency as provided for herein. Accordingly, in some embodiments, a first step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a first time period after which a second step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a second time period, wherein the second step-up dose is not of the same group J, K, or L as the first step-up dose and wherein the first and second time periods may be the same or different. In embodiments comprising a third step-up dose, a third step-up dose selected from J, K, or L as provided for herein is administered at a frequency selected from daily, every other day, or weekly for a third time period, wherein the first, second, and third step-up doses are not of the same group J, K, or L and wherein the first, second, and third time periods may be the same or different.
[0249] In some embodiments, the treatment dose of the glucocorticoid is from about 8 mg to about 50 mg, or any value or range in-between. In some embodiments, the treatment dose of the glucocorticoid is 8 mg. In some embodiments, the treatment dose of the glucocorticoid is 10 mg. In some embodiments, the treatment dose of the glucocorticoid is 12 mg. In some embodiments, the treatment dose of the glucocorticoid is 14 mg. In some embodiments, the treatment dose of the glucocorticoid is 16 mg. In some embodiments, the treatment dose of the glucocorticoid is 18 mg. In some embodiments, the treatment dose of the glucocorticoid is 20 mg. In some embodiments, the treatment dose of the glucocorticoid is 25 mg. In some embodiments, the treatment dose of the glucocorticoid is 30 mg. In some embodiments, the treatment dose of the glucocorticoid is 35 mg. In some embodiments, the treatment dose of the glucocorticoid is 40 mg. In some embodiments, the treatment dose of the glucocorticoid is 45 mg. In some embodiments, the treatment dose of the glucocorticoid is 50 mg.
[0250] In some embodiments, the treatment dose of the glucocorticoid is administered daily. In some embodiments, the treatment dose of the glucocorticoid is administered weekly. In some embodiments, the treatment dose of the glucocorticoid is administered biweekly. In some embodiments, the treatment dose of the glucocorticoid is administered once every three weeks. In some embodiments, the treatment dose of the glucocorticoid is administered once every four weeks. In some embodiments, the treatment dose of the glucocorticoid is administered once a month. The frequency of administration of the treatment dose of the glucocorticoid may be altered as necessary to achieve the desired effect. For example, the treatment dose of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a certain period of time, after which time the treatment dose of the glucocorticoid may be administered daily, weekly, biweekly, once every three weeks, once every four weeks, once every month, or any frequency in-between any of the frequencies recited for a next period of time.
Timing of administration for pre-treatment and co-treatment regimens
[0251] In some embodiments, the components of the pre-treatment regimen or the co-treatment regimen are administered prior to the start of administering the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD). In some embodiments, the components of the pre-treatment regimen or the co-treatment regimen are administered concurrently with the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD).
[0252] In embodiments wherein the therapeutically effective amounts of the glucocorticoid, antihistamine, and antipyretic are administered concurrently with the therapeutically effective amount of the BCMA x CD3 bispecific antibody, the therapeutically effective amount of the anti-CD38 antibody, and the therapeutically effective amount of the immunomodulatory drug (IMiD), the glucocorticoid, antihistamine, and antipyretic may be administered prior to, at the same time as, or after the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered prior to the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered at the same time as the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered after the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. [0253] In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered about 1 minute to about 360 minutes, prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD, or any value or range in-between. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 15 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 20 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 25 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 30 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 35 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 40 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 45 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 50 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 55 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 60 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 75 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 90 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 105 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 120 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 135 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 150 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 165 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD. In some embodiments, the glucocorticoid, antihistamine, and antipyretic are administered 180 minutes prior to the administration of the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD.
[0254] In some embodiments, the glucocorticoid is administered at a certain time prior to the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered about 1 minute to about 240 minutes, prior to the administration of the anti-CD38 antibody, or any value or range in-between. In some embodiments, the glucocorticoid is administered 15 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 20 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 25 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 30 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 35 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 40 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 45 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 50 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 55 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 60 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 75 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 90 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 105 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 120 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 135 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 150 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 165 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 180 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 210 minutes prior to the administration of the anti-CD38 antibody. In some embodiments, the glucocorticoid is administered 240 minutes prior to the administration of the anti-CD38 antibody.
Combination treatments with the anti-CD3 antibody
[0255] In some embodiments, the therapeutically effective amount of the anti-CD3 antibody is administered alone. It is to be understood that “alone” does not necessarily connote that the anti- CD3 antibody is administered as a monotherapy, but rather that no other drug is administered simultaneously with, or in the same composition as the anti-CD3 antibody. In some embodiments, the anti-CD3 antibody is administered in combination with a substance that reduces the injection volume of a subcutaneous administration of the anti-CD3 antibody. In some embodiments, the substance is a recombinant hyaluronidase. In some embodiments, the recombinant hyaluronidase is a recombinant human hyaluronidase PH20 (rHuPH20). In some embodiments, the hyaluronidase comprises an amino acid sequence of SEQ ID NO: 5. In some embodiments, the anti-CD3 antibody is administered in combination with a recombinant hyaluronidase as provided for herein. In some embodiments, the anti-CD3 antibody is administered subcutaneously in combination with a recombinant hyaluronidase as provided for herein. In some embodiments, the anti-CD3 antibody is administered in combination with a recombinant hyaluronidase comprising an amino acid sequence of SEQ ID NO: 5. In some embodiments, the anti-CD3 antibody is administered subcutaneously in combination with a recombinant hyaluronidase comprising an amino acid sequence of SEQ ID NO: 5.
[0256] The hyaluronidase may be administered at any concentration necessary to achieve the desired effect. In some embodiments, the hyaluronidase is administered at a concentration of about 10,000 U to about 50,000 U, or any value or range in-between. In some embodiments, the hyaluronidase is administered at a concentration of 10,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 20,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 30,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 40,000 U. In some embodiments, the hyaluronidase is administered at a concentration of 50,000 U.
Glucocorticoids, antihistamines, and antipyretics
[0257] Glucocorticoids are a class of cortidosteroids which are part of the feedback mechanism of the immune system and act to reduce certain aspects of immune function, such as inflammation. Glucocorticoids are known in the art, and any appropriate glucocorticoid may be used in any of the embodiments provided herein. Non-limiting examples of glucocorticoids include cortisol, cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, deflazacort, fludrocortisone acetate, deoxycorticosterone acetate, aldosterone, and beclometasone. In some embodiments, the glucocorticoid is dexamethasone.
[0258] Antihistamines are a class of drugs that suppress histamine-induced swelling and vasodilation. Antihistamines are known in the art, and any appropriate antihistamine may be used in any of the embodiments provided herein. Non-limiting examples of antihistamines include acrivastine, alimemazine, amitriptyline, amoxapine, azelastine, bilastine, bromodiphenhydramine, bromopheniramine, buclizine, carbinoxamine, cetirizine, chlorodiphenhydramine, chlorphemiramine, chlorpromazine, chlorprothixene, chloropyramine, cinnarizine, clemastine, clomipramine, clozapine, cyclizine, cyproheptadine, desloratadine, dexbrompheniramine, dexchlorpheniramine, dimenhydrinate, dimetindene, diphenhydramine, dosulepin, doxepin, doxylamine, ebastine, embramine, fexofenadine, hydroxyzine, imipramine, levocabastine, levocetirizine, levomepromazine, loratadine, maprotiline, meclizine, mianserin, mirtazapine, olanzapine, olopatadine, orphenadrine, periciazine, phenindamine, pheniramine, phenyltoloxamine, promethazine, pyrilamine, quetiapine, rupatadine, trazodone, tripelennamine, and tirprolidine. In some embodiments, the antihistamine is selected from the group including, but not limited to, bromodiphenhydramine, chlorodiphenhydramine, diphenhydramine, orphenadrine, or any diphenhydramine equivalent. In some embodiments, the antihistamine is a diphenhydramine equivalent. In some embodiments, the antihistamine is diphenhydramine.
[0259] Antipyretics are drugs or substances that act to reduce fever. Antipyretics are known in the art, and any appropriate antipyretic may be used in any of the embodiments provided herein. Non-limiting examples of antipyretics include ibuprofen, aspirin, acetaminophen, naproxen, nonsteroidal anti-inflammatory drugs (NSAIDs), salicylates, ketoprofen, flurbiprofen, nimesulide, diclofenac, and celecoxib. In some embodiments, the antipyretic is a NSAID or salicylate. In some embodiments, the antipyretic is selected from the group including, but not limited to, ibuprofen, aspirin, acetaminophen, naproxen, ketoprofen, flurbiprofen, nimesulide, diclofenac, and celecoxib. In some embodiments, the antipyretic is ibuprofen. In some embodiments, the antipyretic is aspirin. In some embodiments, the antipyretic is acetaminophen. In some embodiments, the antipyretic is naproxen.
Exemplary treatment methods
[0260] The following treatment methods are exemplary and are not meant to be limiting in any way. In some embodiments, the method is as provided in table 13 below:
TABLE 13 - Exemplary Treatment Method
Figure imgf000096_0001
Figure imgf000097_0001
a May be elevated to 1500 pg/kg QW or an intermediate dose. b If previously elevated to 1500 pg/kg QW, may be further elevated to 3000 pg/kg Q2W. c May be elevated to 6000 pg/kg Q4W. d Administered with 30,000 units of hyaluronidase.
[0261] In some embodiments, the method comprises a tiered dosing regimen, wherein the teclistamab dose is determined based on a subject weight threshold of 60 kg. In some embodiments, the method is as provided in table 14 below:
TABLE 14 - Exemplary Treatment Method
Figure imgf000097_0002
Figure imgf000098_0001
a May be elevated to 300 mg Q4W if Q2W dose determined safe. b May be elevated to 450 mg Q4W if Q2W dose determined safe. c Administered with 30,000 units of hyaluronidase.
[0262] In some embodiments, the methods further comprise a pre-treatment or a co-treatment regimen. In some embodiments, the pre-treatment or co-treatment regimen is as provided in table 15 below:
TABLE 15 - Exemplary Pre-treatment Method
Figure imgf000098_0002
a Also after Grade 2 and >3 administration related reactions to daratumumab. b Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab. c For subjects > 75 years of age or <75 years of age and BMI < 18.5, dexamethasone may be administered at a dose of 20 mg. d Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab. e Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab.
[0263] In some embodiments, the pre-treatment or co-treatment regimen is as provided in table 16 below:
TABLE 16 - Exemplary Pre-treatment Method
Figure imgf000098_0003
Figure imgf000099_0001
a Also after Grade 2 and >3 administration related reactions to daratumumab. b Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab. c For subjects > 75 years of age or <75 years of age and BMI < 18.5, dexamethasone may be administered at a dose of 20 mg. d Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab. e Also after Grade >2 cytokine release syndrome or administration related reactions to teclistamab.
[0264] In some embodiments, the method comprises the treatment regimen of table 13. In some embodiments, the method comprises the treatment regimen of table 14. In some embodiments, the method comprises the treatment regimen of table 13 and further comprises the pre-treatment or co-treatment regimen of table 15. In some embodiments, the method comprises the treatment regimen of table 13 and further comprises the pre-treatment or co-treatment regimen of table 16. In some embodiments, the method comprises the treatment regimen of table 14 and further comprises the pre-treatment or co-treatment regimen of table 15. In some embodiments, the method comprises the treatment regimen of table 14 and further comprises the pre-treatment or co-treatment regimen of table 16.
Types of Cancer
[0265] In some embodiments, the cancer is a hematological malignancy or a solid tumor.
[0266] In some embodiments, the hematological malignancy is a multiple myeloma, a smoldering multiple myeloma, a monoclonal gammopathy of undetermined significance (MGUS), an acute lymphoblastic leukemia (ALL), a diffuse large B-cell lymphoma (DLBCL), a Burkitt's lymphoma (BL), a follicular lymphoma (FL), a mantle-cell lymphoma (MCL), Waldenstrom’s macroglobulinema, a plasma cell leukemia, a light chain amyloidosis (AL), a precursor B-cell lymphoblastic leukemia, a precursor B-cell lymphoblastic leukemia, an acute myeloid leukemia (AML), a myelodysplastic syndrome (MDS), a chronic lymphocytic leukemia (CLL), a B cell malignancy, a chronic myeloid leukemia (CML), a hairy cell leukemia (HCL), a blastic plasmacytoid dendritic cell neoplasm, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, a marginal zone B-cell lymphoma (MZL), a mucosa-associated lymphatic tissue lymphoma (MALT), plasma cell leukemia , anaplastic large-cell lymphoma (ALCL), leukemia or lymphoma.
[0267] In some embodiments, the hematological malignancy is multiple myeloma. In some embodiments, the multiple myeloma is a newly diagnosed multiple myeloma. In some embodiments, the multiple myeloma is a relapsed or a refractory multiple myeloma (RRMM).
[0268] In some embodiments, the multiple myeloma is a high-risk multiple myeloma. Subjects with high-risk multiple myeloma are known to relapse early and have poor prognosis and outcome. Subjects can be classified as having high-risk multiple myeloma is they have one or more of the following cytogenetic abnormalities: t(4;14)(pl6;q32), t(14; 16)(q32;q23), dell7p, IqAmp, t(4; 14)(p 16;q32) and t( 14; 16)(q32;q23), t(4; 14)(pl 6;q32) and dell7p, t( 14; 16)(q32;q23) and dell7p, or t(4; 14)(pl6;q32), t( 14; 16)(q32;q23) and dell7p.
[0269] In some embodiments, the subject having the high-risk multiple myeloma has one or more chromosomal abnormalities comprising: t(4; 14)(pl 6;q32), t( 14; 16)(q32;q23), dell7p, IqAmp, t(4;14)(pl6;q32) and t(14;16)(q32;q23), t(4; 14)(p 16;q32) and dell7p, t(14; 16)(q32;q23) and dell7p; or t(4; 14)(pl 6;q32), t( 14; 16)(q32;q23) and dell7p, or any combination thereof.
[0270] Various qualitative and/or quantitative methods can be used to determine relapse or refractory nature of the disease. Symptoms that can be associated are for example a decline or plateau of the well-being of the patient or re-establishment or worsening of various symptoms associated with solid tumors, and/or the spread of cancerous cells in the body from one location to other organs, tissues or cells.
[0271] The cytogenetic abnormalities can be detected for example by fluorescent in situ hybridization (FISH). In chromosomal translocations, an oncogene is translocated to the IgH region on chromosome 14q32, resulting in dysregulation of these genes. t(4; 14)(pl6;q32) involves translocation of fibroblast growth factor receptor 3 (FGFR3) and multiple myeloma SET domain containing protein (MMSET) (also called WHSC1/NSD2), and t( 14; 16)(q32;q23) involves translocation of the MAF transcription factor C-MAF. Deletion of 17p (dell7p) involves loss of the p53 gene locus. [0272] In some embodiments, the multiple myeloma is relapsed or refractory to treatment with the anti-CD38 antibody, lenalidomide, bortezomib, pomalidomide, carfilzomib, elotuzumab, ixazomib, melphalan or thalidomide, or any combination thereof.
[0273] In some embodiments, the multiple myeloma is relapsed or refractory to treatment with the anti-CD38 antibody. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with lenalidomide. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with bortezomib. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with pomalidomide. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with carfilzomib. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with elotuzumab. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with ixazomib. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with melphalan. In some embodiments, the multiple myeloma is relapsed or refractory to treatment with or thalidomide.
[0274] In some embodiments, the hematological malignancy is the AML.
[0275] In some embodiments, the AML is AML with at least one genetic abnormality. In some embodiments, the AML is AML with multilineage dysplasia. In some embodiments, the AML is therapy-related AML. In some embodiments, the AML is undifferentiated AML. In some embodiments, the AML is AML with minimal maturation. In some embodiments, the AML is AML with maturation. In some embodiments, the AML is acute myelomonocytic leukemia. In some embodiments, the AML is acute monocytic leukemia. In some embodiments, the AML is acute erythroid leukemia. In some embodiments, the AML is acute megakaryoblastic leukemia. In some embodiments, the AML is acute basophilic leukemia. In some embodiments, the AML is acute panmyelosis with fibrosis. In some embodiments, the AML is myeloid sarcoma.
[0276] In some embodiments, the at least one genetic abnormality is a translocation between chromosomes 8 and 21, a translocation or an inversion in chromosome 16, a translocation between chromosomes 15 and 17, changes in chromosome 11, or mutation in fms-related tyrosine kinase 3 (FLT3), nucleophosmin (NPM1), isocitrate dehydrogenase 1(IDH1), isocitrate dehydrogenase 2 (IDH2), DNA (cytosine-5)-methyltransferase 3 (DNMT3A), CCAAT/enhancer binding protein alpha (CEB PA), U2 small nuclear RNA auxiliary factor 1(U2AF1), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), structural maintenance of chromosomes 1A (SMC1A) or structural maintenance of chromosomes 3 (SMC3).
[0277] In some embodiments, the at least one genetic abnormality is the translocation between chromosomes 8 and 21. In some embodiments, the at least one genetic abnormality is the translocation or an inversion in chromosome 16. In some embodiments, the at least one genetic abnormality is the translocation between chromosomes 15 and 17. In some embodiments, the at least one genetic abnormality is changes in chromosome 11. In some embodiments, the at least one genetic abnormality is the mutation in fms-related tyrosine kinase 3 (FLT3). In some embodiments, the at least one genetic abnormality is the mutation in nucleophosmin (NPM1). In some embodiments, the at least one genetic abnormality is the mutation in isocitrate dehydrogenase 1(IDH1). In some embodiments, the at least one genetic abnormality is the mutation in isocitrate dehydrogenase 2 (IDH2). In some embodiments, the at least one genetic abnormality is the mutation in DNA (cytosine-5)-methyl transferase 3 (DNMT3A). In some embodiments, the at least one genetic abnormality is the mutation in CCAAT/enhancer binding protein alpha (CEBPA). In some embodiments, the at least one genetic abnormality is the mutation in U2 small nuclear RNA auxiliary factor 1(U2AF1). In some embodiments, the at least one genetic abnormality is the mutation in enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2). In some embodiments, the at least one genetic abnormality is the mutation in structural maintenance of chromosomes 1A (SMC1A). In some embodiments, the at least one genetic abnormality is the mutation in structural maintenance of chromosomes 3 (SMC3).
[0278] In some embodiments, the at least one genetic abnormality is a translocation t(8; 21)(q22; q22), an inversion inv(16)(pl3; q22), a translocation t( 16; 16)(p 13; q22), a translocation t( 15 ; 17)(q22; ql2), a mutation FLT3-ITD, mutations R132H or R100Q/R104V/F108L/R119Q/I130V in IDH1 or mutations R140Q or R172 in IDH2.
[0279] In some embodiments, the at least one genetic abnormality is the translocation t(8; 21)(q22; q22). In some embodiments, the at least one genetic abnormality is the inversion inv(16)(pl3; q22). In some embodiments, the at least one genetic abnormality is the translocation t( 16; 16)(p 13 ; q22). In some embodiments, the at least one genetic abnormality is the translocation t( 15; 17)(q22; ql2). In some embodiments, the at least one genetic abnormality is the mutation FLT3-ITD. In some embodiments, the at least one genetic abnormality is the mutation R132H in IDH1. In some embodiments, the at least one genetic abnormality is the mutation R100Q/R104V/F108L/R119Q/I130V in IDH1. In some embodiments, the at least one genetic abnormality is the mutation R140Q in IDH2. In some embodiments, the at least one genetic abnormality is the mutation R172 in IDH2.
[0280] In some embodiments, the hematological malignancy is the ALL.
[0281] In some embodiments, the ALL is B-cell lineage ALL, T-cell lineage ALL, adult ALL or pediatric ALL.
[0282] In some embodiments, the ALL is B-cell lineage ALL. In some embodiments, the ALL is T-cell lineage ALL. In some embodiments, the ALL is adult ALL. In some embodiments, the ALL is pediatric ALL.
[0283] In some embodiments, the subject with ALL has a Philadelphia chromosome or is resistant or has acquired resistance to treatment with a BCR-ABL kinase inhibitor.
[0284] In some embodiments, the subject with ALL has the Philadelphia chromosome. In some embodiments, the subject with ALL is resistant or has acquired resistance to treatment with a BCR-ABL kinase inhibitor.
[0285] The Ph chromosome is present in about 20% of adults with ALL and a small percentage of children with ALL and is associated with poor prognosis. At a time of relapse, patients with Ph-i- positive ALL may be on tyrosine kinase inhibitor (TKI) regimen and may have therefore become resistant to the TKI. The anti-CD38 antibodies may thus be administered to a subject who has become resistant to selective or partially selective BCR-ABL inhibitors. Exemplary BCR-ABL inhibitors are for example imatinib, dasatinib, nilotinib, bosutinib, ponatinib, bafetinib, saracatinib, tozasertib or danusertib.
[0286] Other chromosomal rearrangements identified in B-lineage ALL patients are t(v;l lq23) (MLL rearranged), t(l ; 19)(q23;pl3.3); TCF3-PBX1 (E2A-PBX1), t(12;21)(pl3;q22); ETV6- RUNX1 (TEL-AML1) and t(5; 14)(q31 ;q32); IL3-IGH.
[0287] In some embodiments, the subject has ALL with t(v;l lq23) (MLL rearranged), t(l;19)(q23;pl3.3); TCF3-PBX1 (E2A-PBX1), t(12;21)(pl3;q22); ETV6-RUNX1 (TEL-AML1) or t(5; 14)(q31 ;q32); IL3-IGH chromosomal rearrangement. [0288] Chromosomal rearrangements can be identified using well known methods, for example fluorescent in situ hybridization, karyotyping, pulsed field gel electrophoresis, or sequencing.
[0289] In some embodiments, the hematological malignancy is the smoldering multiple myeloma. In some embodiments, the hematological malignancy is the MGUS. In some embodiments, the hematological malignancy is the ALL. In some embodiments, the hematological malignancy is the DLBLC. In some embodiments, the hematological malignancy is the BL. In some embodiments, the hematological malignancy is the FL. In some embodiments, the hematological malignancy is the MCL. In some embodiments, the hematological malignancy is Waldenstrom’s macroglobulinema. In some embodiments, the hematological malignancy is the plasma cell leukemia. In some embodiments, the hematological malignancy is the AL. In some embodiments, the hematological malignancy is the precursor B- cell lymphoblastic leukemia. In some embodiments, the hematological malignancy is the precursor B-cell lymphoblastic leukemia. In some embodiments, the hematological malignancy is the myelodysplastic syndrome (MDS). In some embodiments, the hematological malignancy is the CLL. In some embodiments, the hematological malignancy is the B cell malignancy. In some embodiments, the hematological malignancy is the CML. In some embodiments, the hematological malignancy is the HCL. In some embodiments, the hematological malignancy is the blastic plasmacytoid dendritic cell neoplasm. In some embodiments, the hematological malignancy is Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is non-Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is the MZL. In some embodiments, the hematological malignancy is the MALT. In some embodiments, the hematological malignancy is the plasma cell leukemia. In some embodiments, the hematological malignancy is the ALCL. In some embodiments, the hematological malignancy is leukemia. In some embodiments, the hematological malignancy is lymphoma.
Subject population
[0290] In some embodiments, the subject is an adult 18 years of age or older. In some embodiments, the subject is about 18 to about 100 years old, about 19 to 100 years old, about 20 to 100 years old, about 25 to 100 years old, about 30 to 100 years old, about 35 to 100 years old, about 40 to 100 years old, about 45 to 100 years old, about 50 to 100 years old, about 60 to 100 years old, about 70 to 100 years old, about 80 to 100 years old, about 90 to 100 years old, or any age or age range in-between. In some embodiments, the subject is greater than 100 years old. In some embodiments, the subject is about 18 to about 100 years old, about 18 to about 90 years old, about 18 to about 80 years old, about 18 to about 70 years old, about 18 to about 60 years old, about 18 to about 50 years old, about 18 to about 45 years old, about 18 to about 40 years old, about 18 to about 35 years old, about 18 to about 30 years old, about 18 to about 25 years old, about 18 to about 20 years old, about 18 to about 19 years old, or any age or age range inbetween. In some embodiments, the subject is 18 years of age or older.
[0291] In some embodiments, the subject is an adolescent age 12 to 17. In some embodiments, the subject is younger than 18 years old. In some embodiments, the subject is 12. In some embodiments, the subject is 13. In some embodiments, the subject is 14. In some embodiments, the subject is 15. In some embodiments, the subject is 16. In some embodiments, the subject is 16.
[0292] In some embodiments, the subject has an initial diagnosis of multiple myeloma according to the International Myeloma Working Group (IMWG) diagnostic criteria.
[0293] In some embodiments, the subject has newly diagnosed multiple myeloma. In some embodiments, the subject has been previously diagnosed with multiple myeloma. In some embodiments, a subject having been previously diagnosed with multiple myeloma has received 1 to 3 prior lines of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 1 prior line of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 2 prior lines of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma has received 3 prior lines of therapy. In some embodiments, the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI), immunomodulatory drug (IMiD), or a combination thereof. In some embodiments, the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI). In some embodiments, the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received an immunomodulatory drug (IMiD). In some embodiments, the subject having been previously diagnosed with multiple myeloma having received 1 to 3 prior lines of therapy has received a protease inhibitor (PI) and an immunomodulatory drug (IMiD).
[0294] In some embodiments, the subject is newly diagnosed with multiple myeloma and is not considered a candidate for high-dose chemotherapy with autologous stem cell transplantation (ASCT). In some embodiments, the subject is not considered a candidate for high-dose chemotherapy with ASCT due to advanced age. In some embodiments, the subject is not considered a candidate for high-dose chemotherapy with ASCT due to the presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT. In some embodiments, the subject is not considered a candidate for high-dose chemotherapy with ASCT due to deferral of high-dose chemotherapy with ASCT as initial treatment.
[0295] In some embodiments, the subject has measurable disease prior to the start of therapy. In some embodiments, measurable disease is defined as having a Serum M-protein level > l.Og/dL, urine M-protein level of > 200 mg/24 hours, serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio, or a combination thereof. In some embodiments, measurable disease is defined as having a Serum M-protein level > 1.0g/dL. In some embodiments, measurable disease is defined as having a urine M-protein level of > 200 mg/24 hours. In some embodiments, measurable disease is defined as having a serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio. In some embodiments, measurable disease is defined as having a Serum M-protein level > 1.0g/dL, urine M-protein level of > 200 mg/24 hours, and a serum IgG free light chain (FLC) > lOmg/dL and abnormal serum Ig kappa lambda FLC ratio.
IMWG Diagnostic Criteria
[0296] Multiple myeloma is defined as clonal bone marrow plasma cells > 10% or biopsy- proven bony or extramedullary plasmacytoma and at least one of the following two criteria:
[0297] 1. Evidence of end-organ damage, specifically: C: Hypercalcemia; R: Renal insufficiency; A: Anemia; and B: Bone lesions. In some embodiments, hypercalcemia is defined as a serum calcium > 0.25 mmol/L (> Img/dL) higher than the upper limit of normal, or > 2.75mmol/L (> 1 Img/dL). In some embodiments, hypercalcemia is defined as a serum calcium > 0.25 mmol/L (> Img/dL) higher than the upper limit of normal. In some embodiments, hypercalcemia is defined as a serum calcium > 2.75mmol/L (> 1 Img/dL). In some embodiments, renal insufficiency is defined as creatine clearance <40 mL per minute or serum creatine > 177 pmol/L (>2mg/dL). In some embodiments, renal insufficiency is defined as creatine clearance <40 mL per minute. In some embodiments, renal insufficiency is defined as serum creatine > 177 pmol/L (>2mg/dL). In some embodiments, anemia is defined as a hemoglobin value of > 20 g/L below the lower limit of normal, or a hemoglobin value < 100 g/L. In some embodiments, anemia is defined as a hemoglobin value of > 20 g/L below the lower limit of normal. In some embodiments, anemia is defined as a hemoglobin value < 100 g/L. In some embodiments, bone lesions are defined as one or more osteolytic lesions on skeletal radiography, CT, or PET-CT.
[0298] 2. Any one or more of the following biomarkers of malignancy: Clonal bone marrow plasma cell percentage > 60%, involved/uninvolved serum FLC ratio > 100, and > 1 focal lesions on MRI studies.
Outcomes
[0299] In some embodiments, the subject treated by the methods provided for herein has a partial response (PR) or better. In some embodiments, the subject treated by the methods provided for herein has a very good partial response (VGPR) or better. In some embodiments, the subject treated by the methods provided for herein has a complete response (CR) or better. In some embodiments, the subject treated by the methods provided for herein has a stringent complete response (sCR) or better. Unless indicated otherwise herein, PR, VGPR, CR, sCR, stable disease (SD) and progressive disease (PD) are as defined by the IMWG 2016 criteria. IMWG (2016) response criteria are provided in Table A below.
Table A
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
CR=complete response; FLC=free light chain; IMWG=International Myeloma Working Group; M-protein=monoclonal paraprotein; MR=minimal response; PC=plasma cell; PD=progressive disease; PR=partial response; sCR=stringent complete response; SD=stable disease; VGPR=very good partial response a Presence/absence of clonal cells is based upon the kappa/lambda ratio. An abnormal kappa/lambda ratio by immunohistochemistry or immunofluorescence requires a minimum of 100 plasma cells for analysis. An abnormal ratio reflecting presence of an abnormal clone is kappa/lambda of >4:1 or <1:2. b In some cases, it is possible that the original M-protein light chain isotype is still detected on immunofixation but the accompanying heavy-chain component has disappeared; this would not be considered as a CR even though the heavy-chain component is not detectable, because it is possible that the clone evolved to one that secreted only light chains. Thus, if a participant has IgA lambda myeloma, then to qualify as CR there should be no IgA detectable on serum or urine immunofixation; if free lambda is detected without IgA, then it must be accompanied by a different heavy-chain isotype (IgG, IgM, etc). c Clarifications to the criteria for coding progressive disease: bone marrow criteria for progressive disease are to be used only in participants without measurable disease by M-protein and by FLC levels; “25% increase” refers to M-protein, and FLC, and does not refer to bone lesions, or soft-tissue plasmacytomas and the “lowest response value” does not need to be a confirmed value.
* Clarifications to the criteria for coding CR and VGPR in participants in whom the only measurable disease is by sFLC levels: CR in such participants indicates a normal FLC ratio of 0.26 to 1.65 in addition to CR criteria listed above. VGPR in such participants requires a >90% decrease in the difference between involved and uninvolved FLC levels.
[0300] In some embodiments, PR is defined as having a greater than 50% reduction of serum M- protein and reduction in 24 hours urinary M-protein by >90% or to <200 mg/24 hours. In some embodiments, VGPR is defined as having a serum and urine M-protein level detectable by immunofixation but not on electrophoresis or > 90% reduction in serum M-protein plus urine M- protein level <100 mg/24 h. In some embodiments, CR is defined as having a negative immunofixation on serum and urine and disappearance of any soft tissue plasmacytomas and <
5% plasma cells in bone marrow. In some embodiments, sCR is defined as the CR definition as above plus normal FLC ratio and absence of clonal cells in bone marrow by immunohistochemistry or immunofluorescence.
[0301] In some embodiments, the subject treated by the methods provided for herein will have a reduction in BCMA-expressing cells. In some embodiments, the subject treated by the methods provided for herein will have an increase in serum or plasma proteins over time, such as but not limited to cytokines (such as IL-6, IFN-y, IL- 10, and IL-2Ra). In some embodiments, the subject treated by the methods provided for herein will have pharmacodynamics markers indicative of the mechanism of action of daratumumab, such as reduction in CD38+ NK cells or T reg cells in the periphery. In some embodiments, the subject treated by the methods provided for herein will have a reduction in minimal residual disease (MRD) as determined via bone marrow aspirate DNA and next gen sequencing. In some embodiments, the subject treated by the methods provided for herein will have a sustained MRD-negative complete response (CR), wherein the subject is MRD-negative for greater than or equal to 12 months. A subject with sustained MRD-negative CR (>12 months) is a subject with CR or better who sustains MRD-negative status, as determined by NGS with sensitivity of 10’5, for at least 12 months without any examination showing MRD positive status or progressive disease in between.
[0302] In some embodiments, the subject treated by the methods provided for herein will have a greater positive response as compared to other lines of therapy.
[0303] In some embodiments, the subject treated by the methods provided for herein will maintain or have an improvement in the European Organization for Research and Treatment of Cancer Quality of Life Questionaire (EORTC-QLQ-C30).
[0304] In some embodiments, the subject treated by the methods provided for herein will maintain or have an improvement in the Patient-reported Outcomeds Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE).
[0305] In some embodiments, the subject treated by the methods provided for herein will maintain or have improvement in the EuroQol Five Dimension Questionnaire 5-Level (EQ-5D- 5L). [0306] In some embodiments, the subject treated by the methods provided for herein will achieve improvement in one or more symptoms associated with the disease or disorder.
Pharmaceutical compositions and kits
[0307] The invention also provides a pharmaceutical composition comprising a BCMAxCD3 bispecific antibody and an anti-CD38 antibody as described herein. For example, the composition can comprise a BCMA binding domain comprising a VH of SEQ ID NO: 18 and a VL of SEQ ID NO: 19 and a CD3 binding domain comprising the VH of SEQ ID NO: 20 and the VL of SEQ ID NO: 21, and an anti-CD38 antibody comprising a VH of SEQ ID NO: 34 and the VL of SEQ ID NO:35.
[0308] In some embodiments, the pharmaceutical composition comprises the BCMAxCD3 bispecific antibody comprising the HC1 of SEQ ID NO: 22, the LC1 of SEQ ID NO: 23, the HC2 of SEQ ID NO: 24 the LC2 of SEQ ID NO: 25, and the anti-CD38 antibody comprising the HC of SEQ ID NO: 36 and the LC of SEQ ID NO: 37. In some embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises phenylalanine at position 405 and arginine at position 409 in a first heavy chain (HC1) and leucine at position 405 and lysine at position 409 in a second heavy chain (HC2), wherein residue numbering is according to the EU Index. In some embodiments, the BCMAxCD3 bispecific antibody further comprises proline at position 228, alanine at position 234 and alanine at position 235 in both the HC1 and the HC2.
[0309] The disclosure also provides a kit or a combination comprising the BCMAxCD3 bispecific antibody and the anti-CD38 antibody for use in a method of the application.
Methods of generating antibodies used in the methods of the invention
[0310] The antibodies used in the methods of the invention binding specific antigens may be selected de novo from, for example, a phage display library, where the phage is engineered to express human immunoglobulins or portions thereof such as Fabs, single chain antibodies (scFv), or unpaired or paired antibody variable regions (Knappik et al., J Mol Biol 296:57-86, 2000;
Krebs et al., J Immunol Meth 254:67-84, 2001; Vaughan et al., Nature Biotechnology 14:309-14, 1996; Sheets et al., PITAS (USA) 95:6157-62, 1998; Hoogenboom and Winter, J Mol Biol 227:381, 1991; Marks et al., J Mol Biol 222:581, 1991). Phage display libraries expressing antibody heavy and light chain variable regions as fusion proteins with bacteriophage pIX coat protein as described in Shi et al (2010) J. Mol. Biol. 397:385-96 and Int'l Pat. Pub. No. W02009/085462. The antibody libraries may be screened for binding to the desired antigen, such as BCM A and the obtained positive clones may be further characterized and the Fabs isolated from the clone lysates, and subsequently cloned as full-length antibodies. Such phage display methods for isolating human antibodies are established in the art. See for example: U.S. Pat. No. 5,223,409; U.S. Pat. No. 5,403,484; U.S. Pat. No. 5,571,698; U.S. Pat. No. 5,427,908; U.S. Pat. No. 5,580,717; U.S. Pat. No. 5,969,108; U.S. Pat. No. 6,172,197; U.S. Pat. No. 5,885,793; U.S. Pat. No. 6,521,404; U.S. Pat. No. 6,544,731; U.S. Pat. No. 6,555,313; U.S. Pat. No. 6,582,915; and U.S. Pat. No. 6,593,081.
[0311] T cell redirecting bispecific antibodies may be generated in vitro in a cell-free environment by introducing asymmetrical mutations in the CH3 regions of two monospecific homodimeric antibodies and forming the bispecific heterodimeric antibody from two parent monospecific homodimeric antibodies in reducing conditions to allow disulfide bond isomerization according to methods described in Intl.Pat. Publ. No. WO2011/131746. In the methods, two monospecific bivalent antibodies are engineered to have certain substitutions at the CH3 domain that promote heterodimer stability; the antibodies are incubated together under reducing conditions sufficient to allow the cysteines in the hinge region to undergo disulfide bond isomerization; thereby generating the bispecific antibody by Fab arm exchange. The incubation conditions may optimally be restored to non-reducing. Exemplary reducing agents that may be used are 2- mercaptoethylamine (2-MEA), dithiothreitol (DTT), dithioerythritol (DTE), glutathione, tris(2-carboxyethyl)phosphine (TCEP), L-cysteine and betamercaptoethanol, preferably a reducing agent selected from the group consisting of: 2- mercaptoethylamine, dithiothreitol and tris(2-carboxyethyl)phosphine. For example, incubation for at least 90 min at a temperature of at least 20°C in the presence of at least 25 mM 2-MEA or in the presence of at least 0.5 mM dithiothreitol at a pH of from 5-8, for example at pH of 7.0 or at pH of 7.4 may be used.
[0312] Exemplary CH3 mutations that may be used in a first heavy chain and in a second heavy chain of the bispecific antibody are K409R and/or F405L. [0313] Additional CH3 mutations that may be used include technologies such as Duobody® mutations (Genmab), Knob-in-Hole mutations (Genentech), electrostatically-matched mutations (Chugai, Amgen, NovoNordisk, Oncomed), the Strand Exchange Engineered Domain body (SEEDbody) (EMD Serono), and other asymmetric mutations (e.g., Zymeworks).
[0314] Duobody® mutations (Genmab) are disclosed for example in US9150663 and US2014/0303356 and include mutations F405L/K409R, wild-type/F405L_R409K, T350I_K370T_F405L/K409R, K370W/K409R, D399AFGHILMNRSTVWY/K409R, T366ADEFGHILMQVY/K409R, L368ADEGHNRSTVQ/K409AGRH, D399FHKRQ/K409AGRH, F405IKLSTVW/K409AGRH and Y407LWQ/K409AGRH.
[0315] Knob-in-hole mutations are disclosed for example in W01996/027011 and include mutations on the interface of CH3 region in which an amino acid with a small side chain (hole) is introduced into the first CH3 region and an amino acid with a large side chain (knob) is introduced into the second CH3 region, resulting in preferential interaction between the first CH3 region and the second CH3 region. Exemplary CH3 region mutations forming a knob and a hole are T366Y/F405A, T366W/F405W, F405W/Y407A, T394W/Y407T, T394S/Y407A, T366W/T394S, F405W/T394S and T366W/T366S_L368A_Y407V.
[0316] Heavy chain heterodimer formation may be promoted by using electrostatic interactions by substituting positively charged residues on the first CH3 region and negatively charged residues on the second CH3 region as described in US2010/0015133, US2009/0182127, US2010/028637 or US2011/0123532.
[0317] Other asymmetric mutations that can be used to promote heavy chain heterodimerization are L351Y_F405A_Y407V/T394W, T366I_K392M_T394W/F405A_Y407V, T366L_K392M_T394W/F405A_Y407V, L351 Y_Y407A/T366A_K409F, L351Y_Y407A/T366V_K409F, Y407A/T366A_K409F, or T350V_L351Y_F405A_Y407V/T350V_T366L_K392L_T394W as described in US2012/0149876 or US2013/0195849.
[0318] SEEDbody mutations involve substituting select IgG residues with IgA residues to promote heavy chai heterodimerization as described in US20070287170.
I l l [0319] Other exemplary mutations that may be used are R409D_K370E/D399K_E357K, S354C_T366W/Y349C_ T366S_L368A_Y407V, Y349C_T366W/S354C_T366S_L368A_Y407V, T366K/L351D, L351K/Y349E, L351K/Y349D, L351K/L368E, L351Y_Y407A/T366A_K409F, L351Y_Y407A/T366V_K409F, K392D/D399K, K392D/ E356K, K253E_D282K_K322D/D239K_E240K_K292D, K392D_K409D/D356K_D399K as described in W02007/ 147901, WO 2011/143545, WO2013157954, WO2013096291 and US2018/0118849.
[0320] Additional bispecific or multispecific structures that can be used as BCMAxCD3 bispecific antibodies include Dual Variable Domain Immunoglobulins (DVD) (Int. Pat. Publ. No. WO2009/134776; DVDs are full length antibodies comprising the heavy chain having a structure VH1 -linker- VH2-CH and the light chain having the structure VL1 -linker- VL2-CL; linker being optional), structures that include various dimerization domains to connect the two antibody arms with different specificity, such as leucine zipper or collagen dimerization domains (Int. Pat. Publ. No. WO2012/022811, U.S. Pat. No. 5,932,448; U.S. Pat. No. 6,833,441), two or more domain antibodies (dAbs) conjugated together, diabodies, heavy chain only antibodies such as camelid antibodies and engineered camelid antibodies, Dual Targeting (DT)-Ig (GSK/Domantis), Two-in-one Antibody (Genentech), Cross-linked Mabs (Karmanos Cancer Center), mAb2 (F-Star) and CovX-body (CovX/Pfizer), IgG-like Bispecific (InnClone/Eli Filly), Ts2Ab (Medlmmune/AZ) and BsAb (Zymogenetics), HERCUEES (Biogen Idee) and TvAb (Roche), ScFv/Fc Fusions (Academic Institution), SCORPION (Emergent BioSolutions/Trubion, Zymogenetics/BMS), Dual Affinity Retargeting Technology (Fc-DART) (MacroGenics) and Dual(ScFv)2-Fab (National Research Center for Antibody Medicine— China), Dual- Action or Bis-Fab (Genentech), Dock-and-Eock (DNE) (ImmunoMedics), Bivalent Bispecific (Biotecnol) and Fab-Fv (UCB-Celltech). ScFv-, diabody-based, and domain antibodies, include but are not limited to, Bispecific T Cell Engager (BiTE) (Micromet), Tandem Diabody (Tandab) (Affimed), Dual Affinity Retargeting Technology (DART) (MacroGenics), Single-chain Diabody (Academic), TCR-like Antibodies (AIT, ReceptorLogics), Human Serum Albumin ScFv Fusion (Merrimack) and COMBODY (Epigen Biotech), dual targeting nanobodies (Ablynx), dual targeting heavy chain only domain antibodies. Fc engineering of antibodies
[0321] The Fc region of the BCMAxCD3 bispecific antibodies such as bispecific or multispecific antibodies or the anti-CD38 antibodies may comprise at least one substitution in the Fc region that reduces binding of the BCMAxCD3 bispecific antibodies to an activating Fey receptor (FcyR) and/or reduces Fc effector functions such as Clq binding, complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) or phagocytosis (ADCP).
[0322] Fc positions that may be substituted to reduce binding of the Fc to the activating FcyR and subsequently to reduce effector function are substitutions L234A/L235A on IgGl, V234A/G237A/P238S/H268A/V309L/A330S/P331S on IgG2, F234A/L235A on IgG4, S228P/F234A/ L235A on IgG4, N297A on all Ig isotypes, V234A/G237A on IgG2, K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgGl, H268Q/V309L/ A330S/P331S on IgG2, S267E/L328F on IgGl, L234F/L235E/D265A on IgGl, L234A/L235A/G237A/P238S/H268A/A330S/P331S on IgGl, S228P/F234A/L235A/G237A/P238S on IgG4, and S228P/F234A/L235A/G236- deleted/G237A/P238S on IgG4.
[0323] Fc substitutions that may be used to reduce CDC is a K322A substitution.
[0324] Well-known S228P substitution may further be made in IgG4 antibodies to enhance IgG4 stability.
[0325] An exemplary wild-type IgGl comprises an amino acid sequence of SEQ ID NO: 16. An exemplary wild-type IgG4 comprises an amino acid sequence of SEQ ID NO: 17.
[0326] “Antibody-dependent cellular cytotoxicity,” “antibody-dependent cell-mediated cytotoxicity” or “ADCC” is a mechanism for inducing cell death that depends upon the interaction of antibody-coated target cells with effector cells possessing lytic activity, such as natural killer cells (NK), monocytes, macrophages and neutrophils via Fc gamma receptors (FcyR) expressed on effector cells. For example, NK cells express FcyRIIIa, whereas monocytes express FcyRI, FcyRII and FcyRIIIa. ADCC activity of the antibodies may be assessed using an in vitro assay using cells expressing the protein the antibody binds to as target cells and NK cells as effector cells. Cytolysis may be detected by the release of label (e.g., radioactive substrates, fluorescent dyes or natural intracellular proteins) from the lysed cells. In an exemplary assay, target cells are used with a ratio of 1 target cell to 4 effector cells. Target cells are pre-labeled with BATDA and combined with effector cells and the test antibody. The samples are incubated for 2 hours and cell lysis measured by measuring released BATDA into the supernatant. Data is normalized to maximal cytotoxicity with 0.67% Triton X-100 (Sigma Aldrich) and minimal control determined by spontaneous release of BATDA from target cells in the absence of any antibody.
[0327] “Antibody-dependent cellular phagocytosis” (“ADCP”) refers to a mechanism of elimination of antibody-coated target cells by internalization by phagocytic cells, such as macrophages or dendritic cells. ADCP may be evaluated by using monocyte-derived macrophages as effector cells and cells that express the protein the antibody binds to as target cells also engineered to express GFP or another labeled molecule. In an exemplary assay, effector: target cell ratio may be for example 4: 1. Effector cells may be incubated with target cells for 4 hours with or without the antibody of the invention. After incubation, cells may be detached using accutase. Macrophages may be identified with anti-CDl lb and anti-CD14 antibodies coupled to a fluorescent label, and percent phagocytosis may be determined based on % GFP fluorescence in the CD11+CD14+ macrophages using standard methods.
[0328] “Complement-dependent cytotoxicity,” or “CDC,” refers to a mechanism for inducing cell death in which the Fc effector domain of a target-bound antibody binds and activates complement component Clq which in turn activates the complement cascade leading to target cell death. Activation of complement may also result in deposition of complement components on the target cell surface that facilitate CDC by binding complement receptors (e.g., CR3) on leukocytes. CDC of cells may be measured for example by plating Daudi cells at IxlO5 cells/well (50 pE/well) in RPMI-B (RPMI supplemented with 1% BSA), adding 50 pF of test antibodies to the wells at final concentration between 0-100 pg/mE, incubating the reaction for 15 min at room temperature, adding 11 pF of pooled human serum to the wells, and incubation the reaction for 45 min at 37°C. Percentage (%) lysed cells may be detected as % propidium iodide stained cells in FACS assay using standard methods.
[0329] Binding of the antibody to FcyR or FcRn may be assessed on cells engineered to express each receptor using flow cytometry. In an exemplary binding assay, 2xl05 cells per well are seeded in 96-well plate and blocked in BSA Stain Buffer (BD Biosciences, San Jose, USA) for 30 min at 4°C. Cells are incubated with a test antibody on ice for 1.5 hour at 4°C. After being washed twice with BSA stain buffer, the cells are incubated with R-PE labeled anti -human IgG secondary antibody (Jackson Immunoresearch Laboratories) for 45 min at 4°C. The cells are washed twice in stain buffer and then resuspended in 150 pL of Stain Buffer containing 1:200 diluted DRAQ7 live/dead stain (Cell Signaling Technology, Danvers, USA). PE and DRAQ7 signals of the stained cells are detected by Miltenyi MACSQuant flow cytometer (Miltenyi Biotec, Auburn, USA) using B2 and B4 channel, respectively. Live cells are gated on DRAQ7 exclusion and the geometric mean fluorescence signals are determined for at least 10,000 live events collected. FlowJo software (Tree Star) is used for analysis. Data is plotted as the logarithm of antibody concentration versus mean fluorescence signals. Nonlinear regression analysis is performed.
Chimeric antigen receptors (CAR)
[0330] Chimeric antigen receptors (CARs) are genetically engineered receptors. These engineered receptors can be readily inserted into and expressed by immune cells, including T cells in accordance with techniques known in the art. With a CAR, a single receptor can be programmed to both recognize a specific antigen and, when bound to that antigen, activate the immune cell to attack and destroy the cell bearing that antigen. When these antigens exist on tumor cells, an immune cell that expresses the CAR can target and kill the tumor cell.
[0331] CAR typically comprises an extracellular domain that binds the antigen (e.g., prostate neoantigen), an optional linker, a transmembrane domain, and a cytosolic domain comprising a costimulatory domain and/or a signaling domain.
[0332] The extracellular domain of CAR may contain any polypeptide that binds the desired antigen (e.g., prostate neoantigen or B cell maturation antigen (BCMA)). The extracellular domain may comprise a scFv, a portion of an antibody or an alternative scaffold. CARs may also be engineered to bind two or more desired antigens that may be arranged in tandem and separated by linker sequences. For example, one or more domain antibodies, scFvs, llama VHH antibodies or other VH only antibody fragments may be organized in tandem via a linker to provide bispecificity or multispecificity to the CAR. [0333] The transmembrane domain of CAR may be derived from the transmembrane domain of CD8, an alpha, beta or zeta chain of a T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, 0X40, CD2, CD27, LFA-1 (CDI la, CD 18), ICOS (CD278), 4-1 BB (CD 137), 4-1 BBL, GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRFI), CD 160, CDI 9, IL2R beta, IL2R gamma, IL7R a, ITGA1 , VLA1 , CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CDI Id, ITGAE, CD103, ITGAL, CDI la, LFA-1 , ITGAM, CDI lb, ITGAX, CDI 1c, ITGB1 , CD29, ITGB2, CDI 8, LFA-1 , ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1 , CRT AM, Ly9 (CD229), CD160 (BY55), PSGL1 , CD 100 (SEMA4D), SLAMF6 (NTB-A, Lyl08), SLAM (SLAME1 , CD 150, IPO-3), BLAME (SLAME8), SELPLG (CD 162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and/or NKG2C.
[0334] The intracellular costimulatory domain of CAR may be derived from the intracellular domains of one or more co-stimulatory molecules. Co-stimulatory molecules are well-known cell surface molecules other than antigen receptors or Ec receptors that provide a second signal required for efficient activation and function of T lymphocytes upon binding to antigen. Exemplary co-stimulatory domains that can be used in CARs are intracellular domains of 4- IBB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 (0X40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM, and ZAP70.
[0335] The intracellular signaling domain of CAR may be derived from the signaling domains of for example 003^, CD3E, CD22, CD79a, CD66d or CD39. “Intracellular signaling domain,” refers to the part of a CAR polypeptide that participates in transducing the message of effective CAR binding to a target antigen into the interior of the immune effector cell to elicit effector cell function, e.g., activation, cytokine production, proliferation and cytotoxic activity, including the release of cytotoxic factors to the CAR-bound target cell, or other cellular responses elicited following antigen binding to the extracellular CAR domain.
[0336] The optional linker of CAR positioned between the extracellular domain and the transmembrane domain may be a polypeptide of about 2 to 100 amino acids in length. The linker can include or be composed of flexible residues such as glycine and serine so that the adjacent protein domains are free to move relative to one another. Longer linkers may be used when it is desirable to ensure that two adjacent domains do not sterically interfere with one another. Linkers may be cleavable or non-cleavable. Examples of cleavable linkers include 2A linkers (for example T2A), 2A-like linkers or functional equivalents thereof and combinations thereof. The linker may also be derived from a hinge region or portion of the hinge region of any immunoglobulin.
[0337] Exemplary CARs that may be used are for example CAR that contains an extracellular domain that binds the prostate neoantigen of the invention, CD8 transmembrane domain and CD3^ signaling domain. Other exemplary CARs contain an extracellular domain that binds the prostate neoantigen of the invention, CD8 or CD28 transmembrane domain, CD28, 41BB or 0X40 costimulatory domain and CD3^ signaling domain.
[0338] CARs are generated by standard molecular biology techniques. The extracellular domain that binds the desired antigen may be derived from antibodies or their antigen binding fragments generated using the technologies described herein.
Exemplary Embodiments: Treatment Strategies for Newly Diagnosed Multiple Myeloma
[0339] The inventors have developed novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
[0340] Newly diagnosed multiple myeloma patients can be broadly categorized as either “transplant eligible” or “transplant ineligible.” Eligibility for transplant is typically based on age, fitness, and comorbidities. Systemic therapy, typically administered as a triplet or quadruplet drug regimen, is the mainstay of treatment both for transplant-eligible and transplant-ineligible patients. It is used in combination with ASCT for those eligible. DRd (daratumumab, lenalidomide and dexamethasone) or VRd (bortezomib, lenalidomide and dexamethasone) are two key treatment options for newly diagnosed transplant-ineligible patients with multiple myeloma, i.e., standard of care.
[0341] In clinical practice, the treatment of newly diagnosed patients with multiple myeloma is evolving. For certain patients, high-dose therapy with ASCT is not feasible, mainly due to advanced age, comorbidities, or patient frailty. Approximately 40% of all transplant-eligible patients in the US do not receive high-dose therapy and ASCT as initial treatment. This development is driven both by the effectiveness of new induction regimens (e.g., DRd, VRd) without high-dose therapy and ASCT, as well as patient preference, and is expected to continue to increase. Recently reported and ongoing pivotal studies reflect this development and include both ( 1 ) patients who are assessed as being clinically transplant ineligible and (2) patients who are clinically transplant eligible but do not receive transplant as a first treatment option (transplant deferred). Results from several studies have shown similar Overall Survival outcomes for upfront ASCT versus use of a novel agent-based approach without ASCT, suggesting that deferral of ASCT is not associated with adverse survival outcomes and may be a viable option for appropriate candidates. The IFM2009 study showed no OS benefit for participants who were randomized to VRd + early ASCT compared with participants who received VRd + deferred ASCT until after first relapse. Current NCCN guidelines support this approach and indicate that a delayed ASCT after early stem cell collection and storage is an appropriate option (category 1 recommendation: NCCN 2023).
[0342] Despite numerous therapeutic options, multiple myeloma remains incurable for the vast majority of patients. With each successive relapse, symptoms return, quality of life worsens, and duration of response typically decreases. Recent reports suggest that attrition rates between the first and second lines of therapy are as high as 50% in patients with multiple myeloma and achieving the longest possible PFS with the first line of therapy drives OS outcomes. Therefore, there remains a significant and critical unmet need for new therapeutic options directed at alternative mechanisms of action that can better control the disease, provide deeper, more sustained responses, and yield better long-term outcomes including maintenance of health-related quality of life.
[0343] In the field of oncology, even for a drug that already has an established dose in a particular indication, the Food and Drug Administration (FDA) recommends further clinical studies to identify an optimal dose for a new indication; otherwise, patients may be exposed to unreasonable and significant risk, among other potential deficiencies. See, e.g., Optimizing the Dosage of Human Prescription Drugs and Biological Products for the
Treatment of Oncologic Diseases', Draft Guidance for Industry; January 2023). The present inventors have developed novel dosing regimens for BCMAxCD3 bispecific antibodies that provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy. In particular, the present inventors have developed novel dosing regimens for combination therapies comprising teclistamab, daratumumab and lenalidomide, that may provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy.
[0344] Embodiments of the present invention provide novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. As used herein, subjects that are newly diagnosed have a diagnosis of multiple myeloma according to the IMWG diagnostic criteria and have not received any prior therapy for multiple myeloma or smoldering myeloma (but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent). Thus, newly diagnosed subjects do not have relapsed/refractory multiple myeloma (RRMM), i.e., they are not relapsed and/or refractory to prior multiple myeloma therapy (but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent).
[0345] Subjects that are ineligible for ASCT as initial therapy can be ineligible due to (i) advanced age, or (ii) presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT. Subjects that are “not intended” for ASCT as initial therapy are clinically fit enough to undergo ASCT but defer this treatment option.
[0346] According to an embodiment, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy. According to an embodiment, teclistamab, daratumumab and lenalidomide are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant as initial therapy.
[0347] According to certain embodiments, Tec-DR improves PFS (progression-free survival) and/or the rate of sustained MRD-negative CR (>12 months) compared with DRd in subjects with newly diagnosed multiple myeloma who are ineligible or not intended for ASCT as initial therapy.
[0348] According to certain embodiments, teclistamab in combination with daratumumab subcutaneous (SC) and lenalidomide provide an efficacious and safe approach with higher efficacy rates (e.g., mPFS, mOS and MRD negativity rates) compared to the existing standard of care of daratumumab, lenalidomide and dexamethasone (DRd) and/or bortezomib, lenalidomide and dexamethasone (VRd).
[0349] According to certain embodiments of the methods provided herein, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table B below.
Table B - Exemplary Treatment Method
Figure imgf000122_0001
IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous
[0350] According to alternative embodiments of the methods provided herein, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table C below.
Table C - Exemplary Treatment Method
Figure imgf000123_0001
* Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl.
IV=intravenous; Q2W=every 2 weeks; Q4W=every 4 weeks; SC=subcutaneous
[0351] In the regimen shown in Table C, teclistamab is administered after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete. It is believed that a DR lead-in cycle and an early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) may reduce the rate of infections compared to the regimen shown in Table B, while still achieving robust efficacy.
[0352] According to an embodiment, a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, on a dosing schedule comprising sequential 28-day treatment cycles, wherein administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1 , and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3. In certain embodiments, the regimen further comprises, in Cycles 1 and 2 only, orally or intravenously administering dexamethasone to the subject.
[0353] According to another embodiment, a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 2, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on Day 1 or
Day 2), a second step-up dose of 300 pg/kg (e.g., on Day 3 or Day 4), and then weekly
(QW) doses of 1500 pg/kg (e.g., on Days 8 and 15), and in Cycle 3 and all subsequent treatment cycles, subcutaneously administering a monthly
(Q4W) dose of 3000 pg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first
21 days of each 28-day cycle. In certain embodiments, the regimen further comprises, in Cycles 1-2 only, orally or intravenously administering 20 mg of dexamethasone weekly
(QW) (e.g., on Days 1, 8, 15 and 22).
In certain embodiments, the method achieves a decrease in new infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
In certain embodiments, the method achieves a decrease in new grade >3 infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
In certain embodiments, the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria.
In certain embodiments, the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria.
In certain embodiments, the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria.
In certain embodiments, the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. In certain embodiments, the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
In certain embodiments, the method achieves a rate of sustained MRD-negative CR (>12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy.
In certain embodiments, the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
In certain embodiments, the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months.
In certain embodiments, the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
In certain embodiments, the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years.
In certain embodiments, the method achieves a rate of sustained MRD-negative CR (>12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (>12 months) is greater than a reference rate of sustained MRD- negative CR (>12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
In certain embodiments, after about 50 months of treatment, the rate of sustained MRD- negative CR (>12 months) is greater than the reference rate of sustained MRD-negative CR (>12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%.
In certain embodiments, said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8,
15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle; and for the dexamethasone: in Cycle 1 and all subsequent treatment cycles, orally or intravenously administering 40 mg or 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22).
Enumerated Embodiments
[0354] Provided below are enumerated embodiments of the present invention. These embodiments are illustrative only and do not limit the scope of the present disclosure or of the claims attached hereto.
1. A method of treating a cancer in a subject in need thereof, comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
2. The method of embodiment 1, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
3. The method of embodiment 1 or 2, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO:
21.
4. The method of any one of embodiments 1-3, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO:
22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3. 5. The method of any one of embodiments 1-4, wherein the BCMA x CD3 bispecific antibody is teclistamab.
6. The method of any one of embodiments 1-5, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg.
7. The method of embodiment 6, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is selected from 60 pg/kg, 240 pg/kg, 300 pg/kg, 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg.
8. The method of any one of embodiments 1-5, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg.
9. The method of embodiment 8, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is selected from 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, 100 mg, 150 mg, 200 mg, 300 mg, and 450 mg.
10. The method of any one of embodiments 1-9, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily, weekly, biweekly, once every four weeks or monthly.
11. The method of any one of embodiments 1-10, wherein the method further comprises administering a loading dose of the BCMA x CD3 bispecific antibody prior to the therapeutically effective amount of the BCMA x CD3 bispecific antibody.
12. The method of embodiment 11, wherein the loading dose comprises one or more step-up doses.
13. The method of embodiment 11 or 12, wherein the loading dose comprises 1, 2, 3, or more than 3 step up doses. 14. The method of embodiment 12 or 13, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are from about 60 pg/kg to about 6000 pg/kg.
15. The method of embodiment 14, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from 60 pg/kg, 240 pg/kg, or 300 pg/kg.
16. The method of embodiment 12 or 13, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are from about 3 mg to about 600 mg.
17. The method of embodiment 16, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg.
18. The method of embodiment 11 or 12 wherein the loading dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily every other day, or weekly.
19. The method of any one of embodiments 12 to 17, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are administered at a frequency selected from daily, every other day, or weekly.
20. The method of any one of embodiments 1-19, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
21. The method of any one of embodiments 1-20, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35. 22. The method of any one of embodiments 1-21, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
23. The method of any one of embodiments 1-22, wherein the anti-CD38 antibody is daratumumab.
24. The method of any one of embodiments 1-23, wherein the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg.
25. The method of any one of embodiments 1-24, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
26. The method of any one of embodiments 1-25, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from daily, weekly, biweekly, once every four weeks, or monthly.
27. The method of any one of embodiments 1-26, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
28. The method of any one of embodiments 1-27, wherein the IMiD is lenalidomide.
29. The method of any one of embodiments 1-28, wherein the therapeutically effective amount of the IMiD is from about 15 mg to about 50 mg.
30. The method of any one of embodiments 1-29, wherein the therapeutically effective amount of the IMiD is about 25 mg.
31. The method of any one of embodiments 1 -30, wherein the effective amount of the IMiD is administered at a frequency selected from daily or weekly.
32. The method of any one of embodiments 1-31, wherein the BCMA x CD3 bispecific antibody is administered subcutaneously. 33. The method of any one of embodiments 1-32, wherein the anti-CD38 antibody is administered subcutaneously.
34. The method of any one of embodiments 1-33, wherein the IMiD is administered orally.
35. The method of any one of embodiments 1-34, wherein the method further comprises administering a pre-treatment to the subject.
36. The method of embodiment 35, wherein the pre-treatment comprises a glucocorticoid, an antihistamine, an antipyretic, or combinations thereof.
37. The method of embodiment 36, wherein the glucocorticoid is dexamethasone, the antihistamine is diphenhydramine, and the antipyretic is acetaminophen.
38. The method of any one of embodiments 35-37, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
39. The method of embodiment 38, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
40. The method of embodiment 38 or 39, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
41. A method of treating a cancer in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x
CD3 bispecific antibody of about 60 pg/kg, 240 pg/kg or 300 pg/kg or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg or 6000 pg/kg or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1200 mg to about 2400 mg; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 15 mg to about 50 mg; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the ImiD are administered to the subject for a time sufficient to treat the cancer.
42. The method of embodiment 41, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
43. The method of embodiment 41 or 42, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO:
21.
44. The method of any one of embodiments 41-43, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO:
22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
45. The method of any one of embodiments 41-44, wherein the BCMA x CD3 bispecific antibody is teclistamab.
46. The method of any one of embodiments 41-45, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from about 60 pg/kg, 240 pg/kg, 300 pg/kg or any combination thereof. 47. The method of any one of embodiments 41-46, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof.
48. The method of any one of embodiments 41 to 47, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are administered at a frequency selected from daily, every other day, or weekly.
49. The method of any one of embodiments 41 to 48, wherein the treatment dose of the BCMA x CD3 bispecific antibody is selected from 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof.
50. The method of any one of embodiments 41 to 48, wherein the treatment dose of the BCMA x CD3 bispecific antibody is selected from 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof.
51. The method of any one of embodiments 41 to 50, wherein the treatment dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
52. The method of any one of embodiments 41-51, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28,
SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively. 53. The method of any one of embodiments 41-52, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
54. The method of any one of embodiments 41-53, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
55. The method of any one of embodiments 41-54, wherein the anti-CD38 antibody is daratumumab.
56. The method of any one of embodiments 41-55, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
57. The method of any one of embodiments 41-56, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
58. The method of any one of embodiments 41-57, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
59. The method of any one of embodiments 41-58, wherein the IMiD is lenalidomide.
60. The method of any one of embodiments 41-59, wherein the therapeutically effective amount of the IMiD is about 25 mg.
61. The method of any one of embodiments 41-60, wherein the therapeutically effective amount of the IMiD is administered at a frequency selected from daily or weekly.
62. The method of any one of embodiments 41-61, wherein the method further comprises administering a pre-treatment to the subject. 63. The method of embodiment 62, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
64. The method of embodiment 63, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
65. The method of embodiment 63 or 64, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
66. A method of treating a cancer in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of 60 pg/kg, 240 pg/kg, or 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg, or any combination thereof at a frequency selected from, daily, every other day, or weekly; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 25 mg at a frequency of daily or weekly; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
67. The method of embodiment 66, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
68. The method of embodiment 66 or 67, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO:
21.
69. The method of any one of embodiments 66-68, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO:
22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
70. The method of any one of embodiments 66-69, wherein the BCMA x CD3 bispecific antibody is teclistamab.
71. The method of any one of embodiments 66-70, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28,
SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
72. The method of any one of embodiments 66-71, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35. 73. The method of any one of embodiments 66-72, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
74. The method of any one of embodiments 66-73, wherein the anti-CD38 antibody is daratumumab.
75. The method of any one of embodiments 66-74, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
76. The method of any one of embodiments 66-75, wherein the IMiD is lenalidomide.
77. The method of any one of embodiments 66-76, wherein the method further comprises administering a pre-treatment to the subject.
78. The method of embodiment 77, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
79. The method of embodiment 78, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
80. The method of embodiment 78 or 79, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
81. A method of treating a cancer in a subject in need thereof, comprising: i) determining the subjects weight and assigning a BCMA x CD3 bispecific antibody treatment tier based on said weight, wherein if the subject’s weight is less than or equal to a predetermined threshold the subject is in tier 1, and wherein if the subject’s weight is greater than a pre-determined threshold the subject is in tier 2. ii) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody; iii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody; iv) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody; and v) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD); wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
82. The method of embodiment 81 , wherein the predetermined weight threshold is selected from the group consisting of 50 kg, 55 kg, 60 kg, 65 kg, or 70 kg.
83. The method of embodiment 81 or 82, wherein the predetermined weight threshold is 60 kg-
84. The method of any one of embodiments 81-83, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
85. The method of any one of embodiments 81-84, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
86. The method of any one of embodiments 81-85, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3. 87. The method of any one of embodiments 81-86, wherein the BCMA x CD3 bispecific antibody is teclistamab.
88. The method of any one of embodiments 81-87, wherein the one or more step-up doses of a BCMA x CD3 bispecific antibody for a subject in tier 1 or tier 2 are selected from 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg, or any combination thereof.
89. The method of embodiment 88, wherein the one or more step-up doses of a BCMA x CD3 bispecific antibody for a subject in tier 1 may or may not be different from the one or more step-up doses of a BCMA x CD3 bispecific antibody for a subject in tier 2.
90. The method of any one of embodiments 81-89, wherein the treatment dose of the BCMA x CD3 bispecific antibody for a subject in tier 1 or tier 2 are selected from 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof.
91. The method of embodiment 90, wherein the treatment dose of a BCMA x CD3 bispecific antibody for a subject in tier 1 may or may not be different from the treatment dose of a BCMA x CD3 bispecific antibody for a subject in tier 2.
92. The method of any one of embodiments 81 to 91 , wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily, every other day, or weekly.
93. The method of any one of embodiments 81 to 92, wherein the treatment dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
94. The method of any one of embodiments 81-93, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ
ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
95. The method of any one of embodiments 81-94, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
96. The method of any one of embodiments 81-95, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
97. The method of any one of embodiments 81-96, wherein the anti-CD38 antibody is daratumumab.
98. The method of any one of embodiments 81-97, wherein the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg.
99. The method of any one of embodiments 81-98, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
100. The method of any one of embodiments 81-99, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
101. The method of any one of embodiments 81-100, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
102. The method of any one of embodiments 81-101, wherein the ImiD is lenalidomide.
103. The method of any one of embodiments 81-102, wherein the therapeutically effective amount of the ImiD is from about 15 mg to about 50 mg. 104. The method of any one of embodiments 81-103, wherein the therapeutically effective amount of the ImiD is about 25 mg.
105. The method of any one of embodiments 81-104, wherein the effective amount of the ImiD is administered at a frequency selected from daily or weekly.
106. The method of any one of embodiments 81-105, wherein the method further comprises administering a pre-treatment to the subject.
107. The method of embodiment 106, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
108. The method of embodiment 107, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
109. The method of embodiment 107 or 108, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
110. The method of any one of embodiments 1-109, wherein the BCMA x CD3 bispecific antibody, anti-CD38 antibody, and ImiD are administered on a 28-day treatment cycle.
111. The method of embodiment 110, wherein the method further comprises treating the subject for 1, 2, 3, 4, 5, 6, 7, or more than 7 treatment cycles. 112. The method of embodiment 110 or 111, wherein the frequency, dose, or a combination thereof of the BCMA x CD3 bispecific antibody, anti-CD38 antibody, ImiD, or combination thereof may or may not be changed between a current and a subsequent treatment cycle.
113. The method of any one of embodiments 1 to 112, wherein the anti-CD38 antibody is administered or provided for administration together with a hyaluronidase.
114. The method of embodiment 113, wherein the hyaluronidase is rHuPH20.
115. The method of embodiment 113 or 114, wherein the hyaluronidase is administered or provided at 30,000 U.
116. The method of any one of embodiments 1-115, wherein the cancer is multiple myeloma.
117. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of teclistamab at 60 pg/kg, 240 pg/kg, 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof, on days 2 and 4 of the start of treatment; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of teclistamab of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, 450 mg or any combination thereof, at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of dartumumab of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of lenalidomide of about 25 mg at a frequency of once per day; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
118. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab of 60 pg/kg, on day 2 of a treatment regimen, and administering subcutaneously to the subject a second step-up dose of 240 pg/kg or 300 pg/kg on day 4 of a treatment regimen; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg, on days 8, 15, and 22 of the first treatment cycle; b) administering subcutaneously to the subject a treatment dose of teclistamab of 720 pg/kg or 1500 pg/kg weekly for each treatment cycle thereafter; wherein the 720 pg/kg weekly dose optionally is elevated to 1500 pg/kg weekly; wherein the 1500 pg/kg weekly dose optionally is elevated to 3000 pg/kg biweekly starting at a third treatment cycle; and wherein the 3000 pg/kg biweekly dose optionally is elevated to 6000 pg/kg once every four weeks starting at a seventh treatment cycle; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
119. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab on day 2 of a first treatment cycle and a second step-up dose of teclistamab on day 4 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the first step-up dose is 3 mg teclistamab and the second step-up dose is 15 mg teclistamab, wherein if the subject weight greater than 60 kg then the first step-up dose is 4 mg teclistamab and the second step-up dose is 24 mg or 25 mg teclistamab; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab, on days 8, 15, and 22 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; b) administering subcutaneously to the subject a treatment dose of teclistamab, weekly for a second treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; c) administering subcutaneously to the subject a treatment dose of teclistamab, biweekly for a third, fourth, fifth, and sixth treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is
300 mg; and d) administering subcutaneously to the subject a treatment dose of teclistamab, once every four weeks for a seventh and subsequent treatment cycles, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, optionally wherein the treatment dose is elevated to 300 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 300 mg, optionally wherein the treatment dose is elevated to 450 mg; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
120. The method of any one of embodiments 117-119, wherein the method further comprises administering a pre-treatment regimen.
121. The method of embodiment 120, wherein the pre-treatment regimen comprises: i) intravenously or orally administering a therapeutically effective amount of dexamethasone, wherein the dexamethasone is administered at 20 mg on Day 1 of a first treatment cycle, 16 mg on days 2, 4, 8, 15, and 22 of a first treatment cycle, and 20 mg or 40 mg weekly during each of a second, third, and fourth treatment cycles; ii) intravenously or orally administering a therapeutically effective amount of diphenhydramine, wherein the diphenhydramine is administered at 25 mg to 50 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab; and iii) intravenously or orally administering a therapeutically effective amount of acetaminophen, wherein the acetaminophen is administered at 650 mg to 1000 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab.
122. The method of embodiment 121, wherein the diphenhydramine and acetaminophen are further administered after any Grade 2 or greater cytokine release syndrome or administration related reaction to the teclistamab.
123. The method of embodiment 121 or 122, wherein the dexamethasone is further administered after any Grade 2, 3, or greater than 3 administration related reaction to the daratumumab, and/or any Grade 2 or greater cytokine release syndrome or administration related reaction to the teclistamab. 124. The method of any one of embodiments 120-123, wherein the pre-treatment regimen further comprises administration of montelukast at 10 mg.
125. The method of any one of embodiments 1-5, wherein the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on Day 1 or Day 2), a second step-up dose of 300 pg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 pg/kg (e.g., on Days 8, 15 and 22), in Cycle 2, subcutaneously administering weekly (QW) doses of 1500 pg/kg (e.g., on Days 1, 8, 15 and 22), and starting in Cycle 3, subcutaneously administering bi-weekly (Q2W) doses of 3000 pg/kg (e.g., on Days 1 and 15); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on
Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering
1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 2 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle.
126. The method of any one of embodiments 1-5, wherein the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on
Day 1 or Day 2), a second step-up dose of 300 pg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 pg/kg (e.g., on Days 8, 15 and 22), in Cycle 2, subcutaneously administering weekly (QW) doses of 1500 pg/kg (e.g., on Days 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering bi-weekly (Q2W) doses of 3000 pg/kg (e.g., on Days 1 and 15), and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering monthly (Q4W) doses of 3000 pg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering
1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 2 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle.
127. The method of embodiment 125 or 126, wherein the regimen further comprises: in Cycles 2-4, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22).
128. The method of any one of embodiments 1-5, wherein the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, on a dosing schedule comprising sequential 28-day treatment cycles, wherein administration of the daratumumab starts in Cycle 1 , administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3.
129. The method of embodiment 128, wherein the regimen further comprises: in Cycles 1 and 2 only, orally or intravenously administering dexamethasone to the subject. 130. The method of any one of embodiments 1-5, 128 or 129, wherein the cancer is multiple myeloma and the method comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 2, subcutaneously administering a first step-up dose of 60 pg/kg (e.g., on Day 1 or Day 2), a second step-up dose of 300 pg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 pg/kg (e.g., on Days 8 and 15), and in Cycle 3 and all subsequent treatment cycles, subcutaneously administering a monthly (Q4W) dose of 3000 pg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on
Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle. 131. The method of embodiment 130, wherein the regimen further comprises: in Cycles 1-2, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22).
132. The method of any of embodiments 128-131, wherein the method achieves a decrease in new infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
133. The method of any of embodiments 128-131, wherein the method achieves a decrease in new grade >3 infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
134. The method of any of embodiments 125-133, wherein the subject has newly diagnosed multiple myeloma and is either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
135. The method of any of embodiments 125-133, wherein the subject has newly diagnosed multiple myeloma and is ineligible for autologous stem cell transplant (ASCT) as initial therapy.
136. The method of any of embodiments 125-133, wherein the subject has newly diagnosed multiple myeloma and is not intended for autologous stem cell transplant (ASCT) as initial therapy. 137. The method of any of embodiments 125-136, wherein the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria.
138. The method of any of embodiments 125-136, wherein the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria.
139. The method of any of embodiments 125-136, wherein the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria.
140. The method of any of embodiments 1-139, wherein the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
141. The method of any of embodiments 1-140, wherein the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
142. The method of any of embodiments 1-141, wherein the method achieves a rate of sustained MRD-negative CR (>12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy. 143. The method of any of embodiments 1-142, wherein the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
144. The method of embodiment 143, wherein the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months.
145. The method of any of embodiments 1-144, wherein the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
146. The method of embodiment 145, wherein the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years.
147. The method of any of embodiments 1-146, wherein the method achieves a rate of sustained MRD-negative CR (>12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (>12 months) is greater than a reference rate of sustained MRD-negative CR (>12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
148. The method of embodiment 147, wherein, after about 50 months of treatment, the rate of sustained MRD-negative CR (>12 months) is greater than the reference rate of sustained MRD-negative CR (>12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%.
149. The method of embodiment, wherein said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle; and for the dexamethasone: in Cycle 1 and all subsequent treatment cycles, orally or intravenously administering 40 mg or 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22).
Examples of Embodiment #125 above are described in Table 19 and Table 26. As described in embodiments #125-127 above, the BCMAxCD3 bispecific antibody (e.g., teclistamab) and daratumumab are administered starting from Cycle 1 , lenalidomide is not started until Cycle 2 and dexamethasone may be administered in Cycles 2-4. As described in embodiments #128-129 above, the BCMAxCD3 bispecific antibody (e.g., teclistamab) is not started until Cycle 2 (and is administered monthly (Q4W) starting from its second treatment cycle, i.e., Cycle 3 of the regimen), daratumumab and lenalidomide are started in Cycle 1 (i.e., there is a DR lead-in in Cycle 1) and dexamethasone may be administered in Cycles 1-2.
EXAMPLES
[0355] The following examples are provided to further describe some of the embodiments disclosed herein. The examples are intended to illustrate, not to limit, the disclosed embodiments.
[0356] General Materials and methods
[0357] Antibodies and reagents
[0358] Anti-BCMA/anti-CD3 antibody JNJ-64007957 (referred to as JNJ-957) (described in W02017031104A1) and daratumumab were made by Janssen Pharmaceuticals. [0359] JNJ-957 (teclistamab) comprises a BCMA binding arm BCMB69 comprising a HC and LC of SEQ ID NOs 22 and 23, respectively, and a CD3 binding arm CD3B219 comprising a HC and a LC of SEQ ID NOs 24 and 25, respectively. The amino acid sequences of the CDRs, VH, VL, HC, and LC of each binding arm of teclistamab are shown in Tables 7a and 7b in the detailed description section of this disclosure.
[0360] Bone marrow and peripheral blood mononuclear cells
[0361] Peripheral blood mononuclear cells (PBMCs) from healthy donors and MM patients, and bone marrow mononuclear cells (BM-MNCs) from MM patient BM aspirates were isolated by Ficoll-Hypaque density-gradient centrifugation.
[0362] Flow cytometric analysis of bone marrow and blood samples from MM patients
[0363] BM-localized MM cells were identified and analysed for cell surface marker expression levels by staining l.OxlO6 cells/mL with HuMax-003 (CD38) FITC (this antibody binds to an epitope distinct from the epitope bound by daratumumab, Janssen Pharmaceuticals), CD138 PE, CD56 PC7, CD45 Krome Orange (all Beckman Coulter), CD269 (BCMA) APC (Biolegend), CD274 (PD-L1) BV421 and CD19 APC-H7 (both Becton Dickinson). BM or PB immune cell subsets were identified and analysed for cell surface marker expression levels by staining l.OxlO6 cells/mL with CD45 Krome Orange, CD56 PC7 (both Beckman Coulter), CD 14 APC- H7, CD 19 APC-H7, CD3 V450, CD4 APC-H7 or PE, CD8 FITC, CD45-RA APC, CD 127 PE.Cy7, CD62L PE, CD274 (PD-1) BV421, CD 16 APC, HLA-DR APC-H7 (all Becton Dickinson) and CD25 PE (Dako) or with CD4 BUV395 (BD Biosciences), CD8 BUV737 (BD Biosciences), PD-1 BV421 (BD Biosciences), TIM-3 BV650 (BD Biosciences), CD3 BV711 (BD Biosciences), CD45RO BV786 (BD Biosciences), CD38 Humab-003-FITC (Janssen), CD45RA PerCP-Cy5.5 (BD Biosciences), HLA-DR PE (BD Biosciences), LAG-3 PE-eF610 (ThermoFisher), CD25 PE-Cy7 (BD Biosciences), CCR7 AF647 (BD Biosciences), CD 127 APC-eF780 (ThermoFisher). All BM samples were analysed within 24 hours from the time the sample was collected.
[0364] Flow cytometry was performed using a 7-laser LSRFORTESSA (Becton Dickinson). Fluorescent labeled beads (CS&T beads, Becton Dickinson) were used daily to monitor the performance of the flow cytometer and verify optical path and stream flow. This procedure enables controlled standardized results and allows the determination of long-term drifts and incidental changes within the flow cytometer. No changes were observed which could affect the results. Compensation beads were used to determine spectral overlap, compensation was automatically calculated using Diva software. Flow cytometry data were analyzed using FACS Diva software.
[0365] Bioluminescence imaging (BLI)-based lysis assay using LUC-transduced MM cell lines
[0366] LUC-transduced MM cell lines were cultured in the presence or absence of pooled BM stromal cells (BMSCs) obtained from newly diagnosed MM patient (n=12) for 16 hours prior to incubation with effector cells (freshly isolated PBMCs from healthy donors) at an effector to target ratio of 9: 1, and serial dilutions of JNJ-957 (0.00256- 4.0 pg/mL) or control antibodies in 96-well flat bottom plates (Greiner-Bio-One) for 48 hours. The survival of LUC+-MM cells was then determined by BLI, 10 minutes after addition of the substrate luciferin (150 pg/mL;
Promega). Lysis of MM cells was determined using the following formula: % lysis = 1- (mean BLI signal in the presence of effector cells and JNJ-957 / mean BLI signal in the presence of effector cells in untreated wells) xl00%.
[0367] To evaluate the effect of in vivo pretreatment of PB MNCs with daratumumab monotherapy on efficacy of JNJ-957, the LUC-transduced MM cell line 4 was also co-cultured with PB MNCs, obtained from MM patients before initiation of daratumumab monotherapy and at the time of best response to daratumumab monotherapy (effector to target ratio of 9:1). The BLI assay was performed as described before.
[0368] Cytogenetic analysis
[0369] Cytogenetic abnormalities were assessed in purified MM cells by fluorescence in situ hybridization (FISH) and single nucleotide polymorphism (SNP) array. High-risk disease was defined by the presence of del(17p), del(lp), ampl(lq), t(4; 14) or t( 14; 16)2.
[0370] Soluble BCMA Assay
[0371] Soluble BCMA (sBCMA) was measured in cell culture supernatants using MSD GOLD™ 96-well Small Spot Streptavidin SECTOR plates (Meso Scale Diagnostics), according to the manufacturer’s recommended protocol.
[0372] Multiplex Cytokine Assay [0373] Cytokines [interferon-gamma (IFN-y), interleukin (IL)-2, IL-6, IL-8, IL- 10, and tumor necrosis factor-alpha (TNF-a)] in the cell culture supernatants were analyzed using V-Plex proinflammatory Panel 1 Human Kit (Meso Scale Diagnostics), according to the manufacturer’s protocol.
[0374] Statistics
[0375] Comparisons between variables were performed using two-tailed (paired) Student’s /-test, or Mann-Whitney U test or Wilcoxon matched-pairs signed-rank test in case the data do not follow a normal distribution. Correlations between variables were made using the Spearman’s rank correlation coefficient. P-values below 0.05 were considered significant. In case of combinatorial treatment of JNJ-957 and daratumumab, the expected lysis values were calculated to test the null hypothesis that there is only an additive effect between JNJ-957 and daratumumab, using the following formula: % expected lysis = (% lysis with JNJ-957 + % lysis with daratumumab) - (% lysis with JNJ-957 x % lysis with daratumumab), as described before. The null hypothesis of “additive effects” was rejected, if the observed values were significantly higher (P<0.05) than the expected values.
[0376] EXAMPLE 1 - Multi-arm Phase 1 study of Teclistamab with Other Anticancer Therapies in Participants with Multiple Myeloma (MajesTEC-2)
[0377] A phase lb, open label, nonrandomized, multicenter dose-escalation study of teclistamab in combination with subcutaneous (SC) daratumumab and oral lenalidomide administered to adult subjects with multiple myeloma was carried out (Treatment Regimen E). Additional treatment combinations were studied (Treatment Regimens A, B, C, D, F), which included i) teclistamab (SC), daratumumab (SC), and pomalidomide (oral); ii) teclistamab (SC), daratumumab (SC), lenalidomide (oral), and bortezomib (SC) for a 21 day treatment cycle; iii) teclistamab (SC) and nirogacestat (SC); iv) teclistamab (SC) and lenalidomide (oral); and v) teclistamab (SC), daratumumab (SC), lenalidomide (oral), and bortezomib (SC) for a 28 day treatment cycle. The overall aim of the study was to characterize the safety and tolerability of teclistamab combination regimens by assessing the incidence and severity of adverse events (AEs), laboratory values, and the frequency and type of dose-limiting toxicities (DLTs). Safety was monitored throughout the study by a Study Evaluation Team (SET).
[0378] Objectives and Endpoints [0379] Table 17. Objectives and endpoints of the phase lb study of teclistamab (JNJ-957) administered in combination with the various combinations provided for herein for multiple myeloma.
Figure imgf000164_0001
Abbreviations: AE=adverse event; CR=complete response; IMWG=International Myeloma Working Group; mBCAM=membrane BCMA; MRD=minimal residual disease; rHuPH20=recombinant human hyaluronidase PH20 enzyme; PR=partial response; sBCMA=soluble BCMA; sCR=stringent complete response; VGPR=very good partial response [0380] Study Design
[0381] The following treatment combinations were studies:
• Treatment Regimen A - teclistamab + daratumumab + pomalidomide (tec-dara-pom)
• Treatment Regimen B - teclistamab + daratumumab + lenalidomide + bortezomib (tec- dara-len-bor)
• Treatment Regimen C - teclistamab + nirogacestat (tec-niro)
• Treatmetn Regimen D - teclistamab + lenalidomide (tec-len)
• Treatment Regimen E - teclistamab + daratumumab + lenalidomide (tec-dara-len)
• Treatment Regimen F - teclistamab + daratumumab + lenalidomide + bortezomib; 28- day cycle schedule
[0382] For participants in each treatment regimen, teclistamab is administered at a starting dose of 720 pg/kg SC weekly, 1 dose level below the monotherapy recommended phase 2 dose (RP2D) (i.e., 1500 pg/kg SC weekly) as determined based on preliminary data from Study 64007957MMY1001 (NCT03145181).
[0383] In addition to the weight-based dose regimen stated above, a fixed dose regimen was also explored for Regimens D, E, and F. The fixed dose regimen will be separated into 2 tiers by body weight: participants < 60 kg and those > 60 kg.
[0384] Subject Population
[0385] The study was conducted on subjects > 18 years of age with a documented initial diagnosis of multiple myeloma according to IMWG diagnostic criteria. Additional Treatment Regimen specific population requirements were required. For brevity, the following study details, treatment regimens, and results will be limited to Treatment Regimen E, tec-dara-len
[0386] The inclusion and exclusion criteria for enrolling subjects in this study are described below.
[0387] Inclusion criteria
[0388] Each participant must have satisfied all of the following criteria to be enrolled in the study: 1. Be > 18 years of age.
2. Sign an informed consent form (ICF) indicating that he or she understands the purpose of, and procedures required for, the study and is willing to participate in the study including the requirement to provide information during the posttreatment follow-up period. Consent must be obtained prior to the initiation of any study related tests or proceudres that are not part of standard of care for the participant’s disease.
3. Have documented initial diagnosis of multiple myeloma according to IMWG diagnostic criteria.
4. Meet Treatment Regimen E specific requirements as follows: a. Participant has newly diagnosed multiple myeloma or if previously treated has received 1 to 3 prior lines of therapy, including exposure to a PI and an IMiD.
5. Have measurable disease at screening as defined by at least one of the following: a. Serum M-protein level > 1.0 g/dL; or b. Urine M-protein level > 200 mg/24 hours; or c. Light chain multiple myeloma; Serum Ig free light chain (FLC) > 10 mg/dL and abnormal serum Ig kappa lambda FLC ratio.
6. Have an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 of 1 at screening and immediately before the start of study treatment administration.
7. Have clinical laboratory values meeting the following criteria during the Screening Period:
[0389] Table 18. Criteria for clinical laboratory values
Figure imgf000166_0001
Figure imgf000167_0001
Abbreviations: ANC=absolute neutrophil count; ALT=alanine aminotransferase; AST=aspartate aminotransferase; G-CSF=granulocyte colony stimulating factor; GM-CSF=granulocyte-macrophage colonystimulating factor; peg-G-CSF=pegylated granulocyte colony-stimulating factor; RBC=red blood cell;
ULN=upper limit of normal.
8. Women of childbearing potential must have a negative serum (P-human chorionic gonadotropin [P-hCG]) pregnancy test at screening and a negative urine or serum pregnancy test within 24 hours before the start of study treatment administration and must agree to further serum or urine pregnancy tests during the study.
9. Women must be either of the following: a. Not of childbearing potential and b. Of childbearing potential and i. Practicing true abstinence; ii. Or have a sole partner who is vasectomized; iii. Or practicing at least 1 highly effective user-independent method of contraception. If hormonal contraception is used (e.g., oral estrogen/progestin), a male or female condom with or without spermicide (e.g., spermicidal foam/gel/cream/suppository) must also be used.
1. Women of childbearing potential must be on 2 methods of reliable birth control simultaneously while receiving treatment and until 100 days after last dose of treatment: one highly effective form of contraception (tubal ligation, intrauterine device, hormonal [oral, injectable, transdermal patches, vaginal rings, or implants], or partner’s vasectomy), and 1 additional effective contraceptive method (male latex or synthetic condom, diaphragm, or cervical cap) iv. Agree to pregnancy testing (serum or urine) within 100 days after the last dose of study treatment.
Note: If a woman becomes of childbearing potential after start of the study the woman must comply with point (b.) as described above. If a participant’s reproductive status is questionable, additional evaluation should be considered.
10. Men must wear a condom (with or without spermicidal foam/gel/cream/suppository) when engaging in any activities that allow for passage of ejaculate to another person during this study and for 100 days after the last dose of study treatment. His female partner, if of childbearing potential must also be practicing a highly effective method of contraception. If the male participant is vasectomized, he still must wear a condom (with or without spermicidal foam/gel/film/cream/suppository), but his female partner is not required to use contraception.
11. Women must agree not to donate eggs (ova, oocytes) for the purpose of assisted reproduction during the study and for at least 100 days after the last dose of study treatment.
12. Men must agree not to donate sperm or semen for reproduction during the study and for a minimum of 100 days after receiving the last dose of study treatment.
13. Be willing to adhere to the lifestyle restrictions specified in this protocol, including adherence to the applicable IMiD global PPP or local PPP/REMS program.
[0390] Exclusion Criteria
[0391] Any potential subject who meets any of the following criteria will be excluded from participating in the study:
1. Prior treatment with any therapy that targets BCMA.
2. Prior antitumor therapy as follows, in the specified time frame prior to the first dose of the study treatment: a. Targeted therapy, epigenetic therapy, or treatment with an investigational drug or an invasive investigational medical device within 21 days or at least 5 half-lives, whichever is less. b. Gene-modified adoptive cell therapy (e.g., CAR modified T cells, NK cells) within 3 months. c. mAh treatment for multiple myeloma treatment within 21 days. d. Cytotoxic therapy within 21 days. e. PI therapy within 14 days. f. Immunomodulatory agent therapy within 7 days. g. Radiotherapy within 14 days. However, if palliative focal radiation is used, the participant is eligible irrespective of the end date of radiotherapy.
3. Live, attenuated vaccine within 30 days before the first dose of study treatment.
4. Non-hematologic toxicity from prior anticancer therapy that has not resolved to baseline level or to Grade < 1 (except alopecia [any grade] or peripheral neuropathy Grade < 3).
5. Received a cumulative dose of corticosteroids equivalent to > 140 mg of prednisone within the 14-day period before the start of study treatment administration.
6. Received either of the following: a. An allogenic SCT within 6 months before the first dose of study treatment. Participants who received an allogenic transplant must be off all immunosuppressive medications during the 6 weeks before the start of study treatment administration without signs of graft- versus-host disease. b. An autologous SCT within 12 weeks before the start of study treatment administration.
7. Active central nervous system (CNS) involvement or exhibition of clinical signs of meningeal involvement of multiple myeloma. If either is suspected, brain magnetic resonance imaging (MRI) and lumbar cytology are required. 8. Active plasma cell leukemia (> 2.0 x 109/L plasma cells by standard differential), Waldenstrom’s macroglobulinemia, POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes), or primary amyloid light chain amyloidosis.
9. Known to be seropositive for human immunodeficiency virus.
10. Seropositive for HBV, defined by a positive test for hepatitis B surface antigen (HBsAg). Participants with resolved infection (i.e., participants who are HBsAg negative but positive for hepatitis B core antibody [anti-HBc] and/or positive for hepatitis B surface antibody [anti-HBs]) must be screened using real-time polymerase chain reaction (PCR) measurement of HBV DNA levels. Those who are PCR positive will be excluded. EXCEPTION: Participants with serologic findings suggestive of HBV vaccination (anti-HBs positivity as the only serologic marker) and a known history of prior HBV vaccination do not need to be tested for HBV DNA by PCR.
11. Active hepatitis C infection as measured by positive hepatitis C virus (HCV)-RNA testing. Participants with a history of HCV antibody positivity must undergo HCV-RNA testing.
12. Known allergies, hypersensitivity, or intolerance to any study treatment or their excipients (refer to the applicable Investigator’s Brochures and package inserts).
13. Any serious underlying medical condition, such as: a. Evidence of serious active viral, bacterial, or uncontrolled systemic fungal infection. b. Active autoimmune disease or a documented history of autoimmune disease, with the exception of vitiligo, resolved childhood atopic dermatitis, and prior Graves’ disease that is currently euthyroid based on clinical symptoms and laboratory testing. c. Disabling psychiatric conditions (e.g., ongoing alcohol or drug abuse), sever dementia, or altered mental status. d. Any other issue that would impair the ability of the participant to receive, absorb, or tolerate the planned treatment at the study site, to understand informed consent, or any condition for which, in the opinion of the investigator, participation would not be in the best interest of the participant (e.g., compromise well-being) or that could prevent, limit, or confound the protocol-specified assessments. 14. History of stroke or seizure within 6 months prior to the first dose of study treatment.
15. Any of the following cardiac conditions: a. New York Heart Association stage III or IV congestive heart failure. b. Myocardial infarction, unstable angina, or coronary artery bypass graft < 6 months prior to enrollment. c. History of clinically significant ventricular arrhythmia or unexplained syncope not believed to be vasovagal in nature or due to dehydration. d. History of sever nonischemic cardiomyopathy.
16. Pregnant or breastfeeding or planning to become pregnant while enrolled in this study or within 100 days after the last dose of study treatment.
17. Planning to father a child while enrolled in this study or within 100 days after the last dose of study treatment.
18. Major surgery within 2 weeks of the first dose of study treatment, or will not have fully recovered from surgery, or has surgery planned during the time the participant is expected to be treated in the study or within 2 weeks after the last dose of study treatment administration. Note: participants with planned surgical procedures to be conducted under local anesthesia may participate.
[0392] Study Interventions
[0393] The treatments were administered in 28-day cycles. Varying doses of Teclistamab (Tec) were administered with a constant dose of daratumumab (dara) and lenalidomide (len).
[0394] For weight based teclistamab dosing, SC step up doses of 60 pg/kg and 240 pg/kg were administered on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 720 pg/kg administered on days 8, 15, and 22 of treatment cycle 1. Teclistamab 720 pg/kg SC was administered weekly for the second and all subsequent treatment cycles. A separate cohort was administered teclistamab at 1500 pg/kg SC on days 8, 15, and 22 of treatment cycle 1, followed by 1500 pg/kg SC weekly for treatment cycle 2 and then 4000 pg/kg SC biweekly or 3000 pg/kg SC biweekly for treatment cycle 3 and each subsequent treatment cycle. Dosing of teclistamab SC optionally can be further modified as determined by the SET. For example, teclistamab SC 720 pg/kg QW may be elevated to 1500 pg/kg QW as determined by the SET, and may be further elevated to, for example, 3000 pg/kg Q2W beginning at treatment cycle 3 and 6000 pg/kg Q4W or 3000 pg/kg Q4W beginning at treatment cycle 7.
[0395] For fixed dosing, subjects are stratified into two cohorts based on weight. Subjects < 60 kg receive a teclistamab SC step up doses of 3 mg and 15 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 100 mg on days 8, 15, and 22 of treatment cycle 1. Teclistamab SC 100 mg is administered weekly for the second treatment cycle, followed by 200 mg Q2W starting at treatment cycle 3 and, if Q2W dosing is deemed safe, 300 mg Q4W starting at treatment cycle 7. Subjects > 60 kg receive a teclistamab SC step up doses of 4 mg and 25 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 150 mg on days 8, 15, and 22 of treatment cycle 1. Tec SC 150 mg is administered weekly for the second treatment cycle, followed by 300 mg Q2W starting at treatment cycle 3 and, if Q2W dosing is deemed safe, 450 mg Q4W starting at treatment cycle 7.
[0396] Daratumumab was administered to all subjects by SC injection at a dose of 1800 mg with 30000 units hyaluronidase to decrease injection volume required. Daratumumab SC was administered weekly in treatment cycles 1 and 2, Q2W in treatment cycles 3-6, and Q4W thereafter.
[0397] Lenalidomide was administered orally at 25 mg every day for 21 days of a 28-day treatment cycle. Lenalidomide treatment started at treatment cycle 2.
[0398] When daratumumab and teclistamab were administered on the same day, daratumumab was administered first. Step-up dose 1 of teclistamab is administered at least 20 hours after daratumumab SC. The first treatment dose of teclistamab is administered 3 hours after daratumumab. Subsequent treatment doses of teclistamab are administered 1 hour after daratumumab SC.
[0399] The treatment regimen also includes administration of dexamethasone weekly during treatment cycles 2, 3, and 4. Dexamethasone may be administered at 40 mg, or if the participant is greater than 75 years of age or less than or equal to 75 years of age and has a BMI less than 18.5 dexamethasone may be administered at a dose of 20 mg. [0400] The treatment also includes required pretreatment medications. The required pretreatment medications include dexamethasone (glucocorticoid), IV or oral. Dexamethasone is administered at 20 mg on day 1 for the first dose of daratumumab SC and 16 mg for all step-up doses and first treatment doses of teclistamab SC. Dexamethasone is administered on day 1 for the first dose of daratumumab SC and after grade 2 administration related reaction (sAAR) and Grade > 3 sARRs related to daratumumab SC. Dexamethasone is also administered after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC. Dexamthasone is administered approximately 1 to 3 hours prior to daratumumab SC or teclistamab SC on days on which daratumumab SC is not administered. Additional required pretreatment medications include diphenhydramine (anti-histamine) or an equivalent and acetaminophen (antipyretic). Diphenhydramine is administered at 25 mg to 50 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC, and after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC. Diphenhydramine is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC.
Acetaminophen is administered at 650 mg to 1000 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC, and after Grade > 2 cytokine release syndrome or sARRs related to teclistamab SC. Acetaminophen is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC. On days when daratumumab SC and teclistamab SC are given together, pretreatment medications should be given prior to daratumumab SC. If there are greater than 4 hours between administration of study drugs, a repeat dose of dexamethasone 8 mg must be given prior to the bispecific antibody.
Preliminary Results (Cut-Off Date for Analysis July 11, 2022)
[0401] The study is ongoing and as of the cut-off fate for analysis, 32 patients treated with teclistamab, daratumumab, and lenalidomide were evaluated.
[0402] Teclistamab SC + Daratumumab SC + oral lenalidomide Cohorts
[0403] SC treatment with teclistamab and daratumumab and oral treatment with lenalidomide was administered in 28-day cycles (with step-up dosing for teclistamab). Preliminary data as of July 11, 2022, included results for 32 subjects. A summary of the dosing cohorts is shown in Table 19 below. TABLE 19: Dosing Cohorts
Figure imgf000174_0001
a. Step-up dosing was used for teclistamab and two step up doses were given within 1 week before the full dose. Step-up doses for the 720 pg/kg cohort were 60 pg/kg and 240 pg/kg. Step-up doses for the 1500 pg/kg cohort were 60 pg/kg and 300 pg/kg. b. with transition to 3000 pg/kg Q2W starting at Cycle 3 c. starting a Cycle 2
[0404] Median age range for the 32 subjects evaluated was 62 years (range, 38-75) and 28 subjects were male (87.5%). All patients had received prior multiple myeloma treatment; median prior LOT was 2 (range, 1-3), and 31.3% were anti-CD38 exposed. A summary of the subjects’ demographics and baseline characteristics is shown in Table 20.
TABLE 20: Demographics
Figure imgf000174_0002
Figure imgf000175_0001
’Exposed to >2 Pls, >2 IMiDs, and an anti-CD38 mAb. bRefractory to >1 PI, >1 IMiD, and an anti-CD38 mAb. cRefractory to >2 Pls, >2 IMiDs, and an anti-CD38 mAb.
Dara, daratumumab; len, lenalidomide; LOT, line of therapy; mAb, monoclonal antibody; tec, teclistamab.
[0405] The combination of teclistamab, daratumumab, and lenalidomide exhibited a safety profile consistent with teclistamab monotherapy or daratumumab + lenalidomide. The most frequent AEs were CRS, neutropenia, and fatigue. Grade 3/4 AEs occurred in 29 (90.6%) of patients. The most common grade 3/4 AEs were neutropenia (68.8%), pneumonia (15.6%) and anemia, febrile neutropenia, and lymphopenia (12.5% each). The majority of grade 3/4 AEs occurred in one patient. There were no ICANS events. Infections occurred in 24 patients (75%). A summary of the AEs for subjects treated with teclistamab, daratumumab, and lenalidomide is shown in Table 21 below.
TABLE 21: AE Summary
Figure imgf000176_0001
[0406] CRS was frequent, but low-grade. All CRS events were grade 1/2, and 95% of the events occurred during cycle 1 treatment doses. Median time to onset was 2 days (range, 1-8) and median duration was 2 days (range, 1-22). A summary of CRS events is provided in Table 22 below.
TABLE 22: CRS summary.
Figure imgf000176_0002
’Graded per ASTCT guidelines bConfounded by ongoing infection. c Amikacin, amoxicillin trihydrate; clavulanate potassium, anakinra, aztreonam, cefepime, cefepime hydrochloride, ceftazidime, gentamicin, meropenem, paracetamol, pethidine, piperacillin sodium, tazobactam sodium, sodium chloride, vancomycin.
ASTCT, American Society for Transplantation and Cellular Therapy; CRS, cytokine release syndrome; dara, daratumumab; len, lenalidomide; tec, teclistamab. [0407] Infections were common, but generally low grade. Infections of any grade occurred in 24 patients (75%). The most common infections were CO VID- 19, pneumonia, and upper respiratory infection. Grade 3/4 infections occurred in 9 patients (28.1%). 3 patients (9.4%) had CO VID-19 pneumonia. 1 patient (3.1%) discontinued due to an AE (COVID-19), considered unrelated to the study drugs. This patient subsequently died due to COVID-19. There were no other deaths related to AEs. The rate of infections for the teclistamab + daratumumab + lenalidomide treatment regimen was similar to teclistamab monotherapy, but the rate of grade 3/4 infections was lower than the monotherapy. A summary of the infection data is provided in Table 23 below.
TABLE 23: Infection Summary
Figure imgf000177_0001
[0408] There were 29 response evaluable subjects. The ORR for patients (13) treated with 720 pg/kg QW SC teclistamab + dartumumab SC 1800 mg + lenalidomide was 100%. The ORR for response evaluable patients (16) treated with 1500 pg/kg QW SC teclistamab + dartumumab SC 1800 mg + lenalidomide was 81.25% (13/16). A summary of the ORRs is shown in Table 24 below.
TABLE 24: ORR Summary
Figure imgf000177_0002
Figure imgf000178_0001
Response based on investigator assessment (using IMWG criteria) among all patients who received >1 study treatment and were followed for >1 month or had >1 response evaluation. a16 of the 19 patients in this dose group were response-evaluable.
CR, complete response; dara, daratumumab; IMWG, International Myeloma Working Group; len, lenalidomide; ORR, overall response rate; PR, partial response; sCR, stringent complete response; tec, teclistamab; VGPR, very good partial response
[0409] Preliminary pharmacokinetic analysis demonstrated that concentrations of teclistamab in combination with daratumumab and lenalidomide were similar to those see with teclistamab monotherapy (FIG. 2). Teclistamab + daratumumab + lenalidomide treatment led to T-cell activation and proinflammatory cytokine production as measured by induction of interleukin-6, soluble interleukin-2Ra, interferon-y, and tumor necrosis factor-a)
[0410] Summary
[0411] Patients were eligible for tec-dara-len if they had received 1-3 prior lines of therapy (LOT), including a proteasome inhibitor and immunomodulatory drug. In this cohort, patients received weekly doses of tec (0.72 or 1.5 mg/kg with step-up dosing) plus the approved schedules of dara 1800 mg + len 25 mg. Responses were investigator assessed per International Myeloma Working Group criteria and adverse events (AEs) by CTCAE v5.0, except for cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which were graded per ASTCT guidelines.
[0412] 32 patients received tec-dara-len (0.72 mg/kg, n=13; 1.5 mg/kg, n=19). At the data cutoff (July 11, 2022), median follow-up was 5.78 months (mo; range, 1.0-10.4) and median treatment duration was 4.98 mo (range, 0.10-10.35). Median age was 62 years (range, 38-75); 87.5% were male. All patients had received prior MM treatment; median prior LOT was 2 (range, 1-3), and 31.3% were anti-CD38 exposed. The most frequent AE was CRS (81.3% [n=26]). All CRS events were grade 1/2, and 95% of the events occurred during cycle 1 treatment doses. Median time to onset was 2 days (range, 1-8) and median duration was 2 days (range, 1-22). No ICANS events were reported. Other frequent AEs (>25.0% across both dose levels) were neutropenia (75.0% [n=24]; grade 3/4: 68.8% [n=22]), fatigue (43.8% [n=14]; grade 3/4: 6.3% [n=2]), diarrhea (37.5% [n=12J; all grade 1/2), insomnia (31.3% [n=10]; grade 3/4: 3.1% [n=l]), cough (28.1% [n=9]; all grade 1/2), hypophosphatemia (25.0% [n=8]; all grade 1/2), and pyrexia (25% [n=8] ; grade 3/4: 6.3% [n=2]). The frequency of febrile neutropenia was 12.5% (n=4). Infections occurred in 24 patients (75.0%; grade 3/4: 28.1% [n=9]). The most common infections were upper respiratory infection (21.9% [n=7]), COVID-19 (21.9% [n=7]), and pneumonia (21.9% [n=7]). Three patients (9.4%) had COVID-19 pneumonia. One patient (3.1%) discontinued due to an AE (COVID- 19) considered unrelated to study drugs; this patient died due to COVID- 19. The overall response rate (ORR) was 13/13 evaluable patients (median follow-up, 8.61 mo) at 0.72 mg/kg and 13/16 evaluable patients (median follow-up was less mature at 4.17 mo) at 1.5 mg/kg. Very good partial response or better was achieved in 12 patients at the 0.72 mg/kg dose and was not mature for the 1.5 mg/kg group. Median time to first response was 1.0 mo (range, 0.7-2.0). Preliminary pharmacokinetic concentrations of tec in combination with dara-len were similar to those seen with tec monotherapy. Tec-dara-len treatment led to proinflammatory cytokine production (induction of interleukin-6, soluble interleukin-2Ra, interferon-y, and tumor necrosis factor-a) and T-cell activation (upregulation of programmed cell death protein- 1 and CD38 on peripheral T cells).
[0413] Tec-dara-len was well tolerated, with a safety profile consistent with tec or dara-len individually. Promising ORR supports the potential for this combination to have enhanced early disease control through the addition of teclistamab. The randomized phase 3 MajesTEC-7 study described in Example 2 will compare tec-dara-len vs the combination of dara, len, and dexamethasone in patients with NDMM ineligible or not intended for autologous stem cell transplant as initial treatment.
[0414] EXAMPLE 2 - A Phase 3 Randomized Study Comparing Teclistamab in Combination with Daratumumab SC and Lenalidomide (Tec-DR) versus Daratumumab SC, Lenalidomide, and Dexamethasone (DRd), and Talquetamab in Combination with Daratumumab SC and Lenalidomide (Tal-DR) versus DRd, in Patients with Newly Diagnoses Multiple Myeloma Who are Either Ineligible or not Intended for Autologous Stem Cell Transplant as Initial Therapy (MajesTEC-7)
[0415] A Phase 3 randomized study comparing teclistamab in combination with daratumumab SC and lenalidomide (Tec-DR) versus daratumumab SC, lenalidomide, and dexamethasone (DRd), and Talquetamab in Combination with Daratumumab SC and Lenalidomide (Tal-DR) versus DRd, will be carried out (NCT05552222). The Tal-DR arm of the study was added after initiation of the safety run-in phase of the study. The overall aim is to compare the efficacy between Tec-DR and DRd, and Tal-DR and DRd, in terms of PFS (progression-free survival, which refers to duration from the date of randomization to either progressive disease or death, whichever comes first) and minimal residual disease (MRD)-negative complete response (CR).
[0416] Objectives and Endpoints
[0417] Table 25: Objectives and endpoints of the phase 3 study.
Figure imgf000180_0001
Figure imgf000181_0001
Abbreviations: AE=adverse event; CR=complete response; IMWG=International Myeloma Working Group; mBCAM=membrane BCMA; MRD=minimal residual disease; rHuPH20=recombinant human hyaluronidase PH20 enzyme; PR=partial response; sBCMA=soluble BCMA; sCR=stringent complete response; VGPR=very good partial response
Study Design
[0418] This is a randomized, open-label, multicenter, Phase 3 study in adult participants with newly diagnosed multiple myeloma according to the IMWG diagnostic criteria who are ineligible or not intended for ASCT as initial therapy. Inclusion Criteria and Exclusion Criteria are provided below.
Inclusion Criteria:
• Have a diagnosis of multiple myeloma according to the International Myeloma Working Group (IMWG) diagnostic criteria
• Be newly diagnosed and not considered a candidate for high-dose chemotherapy with autologous stem cell transplant (ASCT) due to: ineligible due to advanced age OR; ineligible due to the presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT OR; deferral of high-dose chemotherapy with ASCT as initial treatment
• Have an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 to 2
• A female participant must agree not to be pregnant, breastfeeding, or planning to become pregnant while enrolled in this study or within 3 months after the last dose of study treatment
• A male participant must agree not to plan to father a child while enrolled in this study or within 3 months after the last dose of study treatment
Exclusion Criteria: • Received a cumulative dose of systemic corticosteroids equivalent to greater than or equal to (>=) 20 milligrams (mg) of dexamethasone within 14 days before randomization
• Had plasmapheresis within 28 days of randomization
• Had a stroke, transient ischemic attack, or seizure within 6 months prior to randomization
• Known allergies, hypersensitivity, or intolerance to teclistamab excipients
• Known contraindications to the use of daratumumab or lenalidomide per local prescribing information
Initial Study Interventions (as of Nov-2022)
[0419] As described in the section below entitled “Updated Study Design Following Safety Run- In Cohort 1 (as of Oct-2023),” the study interventions described here were eventually modified, after initiation of the safety run-in phase of the study.
[0420] The treatments are administered in 28-day cycles. For Tec-DR cohorts: varying doses of Teclistamab (Tec) are administered with a constant dose of daratumumab (dara), lenalidomide (len), and dexamethasone. For DRd cohorts: constant doses of daratumumab (dara), lenalidomide (len), and dexamethasone are administered.
[0421] For teclistamab fixed dosing, subjects are stratified into two cohorts based on weight. Subjects < 60 kg receive a teclistamab SC step up doses of 3 mg and 15 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 100 mg on days 8, 15, and 22 of treatment cycle 1. Teclistamab SC 100 mg is administered weekly for the second treatment cycle, followed by 200 mg Q2W starting at treatment cycle 3, and 200 mg Q4W starting at treatment cycle 7. Subjects > 60 kg receive a teclistamab SC step up doses of 4 mg and 25 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 150 mg on days 8, 15, and 22 of treatment cycle 1. Tec SC 150 mg is administered weekly for the second treatment cycle, followed by 300 mg Q2W starting at treatment cycle 3, and 300 mg Q4W starting at treatment cycle 7.
[0422] Daratumumab is administered to all subjects by SC injection at a dose of 1800 mg with 30000 units hyaluronidase to decrease injection volume required. Daratumumab SC is administered weekly in treatment cycles 1 and 2, Q2W in treatment cycles 3-6, and Q4W thereafter.
[0423] Lenalidomide is administered orally at 25 mg every day for 21 days of a 28-day treatment cycle. Lenalidomide treatment started at treatment cycle 2.
[0424] For the Tec-DR cohort, dexamethasone is administered orally or intravenously at 20 mg weekly. For the Tec-DR cohort in Safety Run in Cohort 1, dexamethasone is administered during treatment cycles 2, 3, and 4. For the DRd cohort, dexamethasone is administered at 40 mg, or if the participant is greater than 75 years of age or has a BMI less than 18.5 dexamethasone is administered at a dose of 20 mg. For the DRd cohort, dexamethasone is administered weekly for all cycles.
[0425] When daratumumab and teclistamab were administered on the same day, daratumumab was administered first. Step-up dose 1 of teclistamab is administered at least 20 hours after daratumumab SC. The first treatment dose of teclistamab is administered 3 hours after daratumumab. Subsequent treatment doses of teclistamab are administered at least 15 minutes after daratumumab SC.
[0426] The treatment also includes required and optional pretreatment medications. The required pretreatment medications include dexamethasone (glucocorticoid), IV or oral. Dexamethasone is administered at 16 mg on days 1, 2, 4 and 8 of treatment cycle 1. Dexamthasone is administered approximately 1 to 3 hours prior to daratumumab SC or teclistamab SC on days on which daratumumab SC is not administered. Additional required pretreatment medications include diphenhydramine (anti-histamine) or an equivalent and acetaminophen (antipyretic). Diphenhydramine is administered at 25 mg to 50 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC. Diphenhydramine is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC. Acetaminophen is administered at 650 mg to 1000 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC. Acetaminophen is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC. Optionally, montelukast (10 mg) may be administered at the discretion of the examiner prior to administration of daratumumab. On days when daratumumab SC and teclistamab SC are given together, pretreatment medications should be given prior to daratumumab SC. If there are greater than 4 hours between administration of study drugs, a repeat dose of dexamethasone 8 mg must be given prior to the bispecific antibody. A participant treated with Tec-DR who experiences Grade >2 CRS/sARR related to teclistamab or daratumumab SC must receive dexamethasone 16 mg, diphenhydramine 25 to 50 mg or equivalent, and acetaminophen 650 to 1000 mg as pretreatment medication for at least the subsequent dose of the study drug to which the event was related. These event-driven pretreatment medications are not required if already being administered as planned pretreatment described above. Dexamethasone doses scheduled as background treatment for Cycles 2-4 in participants treated with Tec-DR can be used as event-driven pretreatment medication.
Summary of Initial Trial Design (as of Nov-2022)
[0427] Eligible patients will be aged >18 years with NDMM and are either ineligible or not intended for ASCT as initial therapy, have measurable disease, and an ECOG performance status score 0-2. A safety run-in phase for Tec-DR will precede the randomized phase. Approximately 1000 patients will be randomized 1: 1 to receive Tec-DR or DRd. Treatment will continue until progressive disease, death, intolerable toxicity, loss to follow-up, withdrawal of consent, or end of study, whichever occurs first. The dual primary endpoints are PFS and minimal residual disease (MRD)-negative complete response (CR) sustained for at least 12 months. Secondary endpoints include rate of CR or better, overall survival, rate of MRD-negative CR, rate of very good partial response or better, PFS2, patient-reported outcomes, and adverse events (AEs). Response will be assessed using 2016 International Myeloma Working Group criteria. AEs will be graded by Common Terminology Criteria for AEs v5.0, except for cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, which will be graded by American Society for Transplantation and Cellular Therapy guidelines. Results from this trial will provide insights into a possible new treatment regimen (Tec-DR) for patients with NDMM, which may provide improved outcomes.
Updated Study Design Following Safety Run-In Cohort 1 (as of Oct-2023)
[0428] Based on evolving data in the teclistamab clinical development program, it was decided to investigate modified dosing schedules following initiation of the Tec-DR Safety Run-In Cohort 1. As such, a second Tec-DR Safety Run-In Cohort 2 (n=30) will be performed in the current study with a modified schedule of teclistamab that includes a lead-in cycle of DRd, administration of teclistamab starting in Cycle 2 and Q4W dosing starting in Cycle 3. Additionally, a Talquetamab-DR (Tal-DR) regimen will be evaluated in Safety Run-in Cohort 3 (n=30). Subsequently, about 1500 participants (500 per arm) will be randomized 1:1:1 to Tec-DR, Tal-DR. See FIG. 4 for a schematic overview of this updated study design.
[0429] The study treatment that was administered for the Tec-DR Safety Run-In Cohort 1 (Tec- DR SRI 1) is provided in Table 26. Participants enrolled in Safety Run-in Cohort 1 were initially treated with fixed dose teclistamab in combination with DR. Following implementation of a protocol amendment, due to a safety measure, all participants treated with fixed dose teclistamab were transitioned to weight-based teclistamab.
Table 26: Study Treatment Administered - Tec-DR - Safety Run-in Cohort 1
Figure imgf000185_0001
IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous
[0430] For the randomized part of this study, the treatment to be administered to subjects in the DRd arm is provided in Table 27.
Table 27: Study Treatment Administered - DRd - Randomized Part
Figure imgf000185_0002
Figure imgf000186_0001
IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous
* Lenalidomide dose may need to be adjusted for participants with reduced CrCl
** For participants >75 years of age or with BMI <18.5 kg/m2, dexamethasone may be administered at a dose of 20 mg.
[0431] As of 10 April 2023, 26 participants with newly diagnosed multiple myeloma were enrolled in Tec-DR Safety Run-in Cohort 1 of the current study and completed at least 2 cycles of treatment with Tec-DR. The median time from diagnosis to enrollment was 1 month (range, 0.16-4.76 months). The median age of participants at time of consent was 72.5 years (range, 66-84 years), and the majority (n=21, 81%) of participants were 70 years of age or older. The majority (n=22, 85%) of participants were transplant ineligible due to advanced age, with the remaining 15% transplant deferred due to a desire for treatment with a novel therapy. Preliminary efficacy results for the TEC-DR combo are shown in Table 28 below, in particular, the results for "VGPR or better” (VGPR, CR or sCR) and “CR or better” (CR or sCR) response rates at median follow-up (mFU) of 6.5 months.
TABLE 28:
Figure imgf000186_0002
[0432] With regard to the safety profile observed in Tec-DR SRI 1, using a median follow-up of
6.7 months, the rate of any grade infection was 92.3%, the rate of Grade 3+ infections was 38.5% and the rate of Grade 3 or 4 infections was 34.6%. By further optimizing the dosing regimen, it may be possible to decrease the rates of infection, thereby improving the safety profile.
[0433] The study treatment to be administered for the Tec-DR Safety Run-In Cohort 2 (Tec-DR SRI 2), and the Randomized Tec-DR Arm, is provided in Table 29. The aim of the dosing regimen used in Tec-DR SRI 2 is to achieve robust efficacy while improving the safety profile seen in Tec-DR SRI 1, including reduced rates of infections.
Table 29: Tec-DR - Safety Run-In Cohort 2 and Randomized Part
Figure imgf000187_0001
* Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl.
[0434] As shown in Table 29, teclistamab will be given after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete. The early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) was implemented with the aim of reducing the rate of severe infections. The teclistamab SC dose schedule for Safety Run-in Cohort 2 of the study will include 2 step-up doses (0.06 and 0.3 mg/kg) on Days 1 and 3 of Cycle 2 followed by 1.5 mg/kg on Days 8 and 15 of Cycle 2 and 3 mg/kg Q4W starting from Cycle 3.
[0435] The modified dosing regimen of teclistamab to be evaluated in Safety Run-in Cohort 2 is supported by emerging data from participants with relapsed/refractory multiple myeloma treated with teclistamab monotherapy (MajesTEC-1 ; Study 64407564MMY 1001). In updated results from MajesTEC-1 as of the clinical cutoff date of 04 January 2023, 63 responders who received the RP2D dose (1.5 mg/kg QW) switched to Q2W treatment with a median time to switch of 11.3 months (range: 1.0 to 24.7) and 9 of those participants had switched from 1.5 mg/kg Q2W to 1.5 mg/kg Q4W dosing. Forty-nine (77.8%) of the 63 participants did not have progressive disease, with a median follow-up of 12.6 months after schedule change. For those participants who switched to 1.5 mg/kg Q2W dosing, median DOR was not reached, and the 24-month event free rate was 68.7% (95% CI: 53.6 to 79.7).
[0436] Additionally, in the MajesTEC-1 study, among those who stayed on treatment for at least 12 months, patients who switched to 1.5 mg/kg Q2W by 12 months experienced a lower incidence of Grade >3 treatment-emergent infections between 12 to 18 months of treatment than those who remained on 1.5 mg/kg QW dosing by 12 months (15.6% vs 33.3%).
[0437] Using a population PK model based on teclistamab clinical data, it is estimated that the 3 mg/kg Q4W dosing regimen will have a lower exposure (concentration-time profile) than 1.5 mg/kg QW at steady state, which has been associated with durable responses. It is also estimated that the 3 mg/kg Q4W dosing regimen will achieve comparable PK parameters at steady state (Ctrough, Cmax, and AUC) to that of 1.5 mg/kg Q2W dosing after switch from 1.5 mg/kg QW. Additionally, it is estimated that the modified Q4W regimen will resemble the exposure (concentration-time profile) of the 0.72 mg/kg QW dose, which has shown robust efficacy and good safety in combination with daratumumab and lenalidomide in MajesTEC-2 (Study 64007957MMY1004).

Claims

Claims
1. A method of treating a cancer in a subject in need thereof, comprising administering a therapeutically effective amount of a BCMA x CD3 bispecific antibody, a therapeutically effective amount of an anti-CD38 antibody, and a therapeutically effective amount of an immunomodulatory drug (IMiD) to the subject for a time sufficient to treat the cancer.
2. The method of claim 1, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
3. The method of claim 1 or 2, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
4. The method of any one of claims 1-3, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
5. The method of any one of claims 1-4, wherein the BCMA x CD3 bispecific antibody is teclistamab.
6. The method of any one of claims 1-5, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 60 pg/kg to about 6000 pg/kg.
7. The method of claim 6, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is selected from 60 pg/kg, 240 pg/kg, 300 pg/kg, 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg.
8. The method of any one of claims 1-5, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is from about 3 mg to about 600 mg.
9. The method of claim 8, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is selected from 3 mg, 4 mg, 15 mg, 25 mg, 100 mg, 150 mg, 200 mg, 300 mg, and 450 mg.
10. The method of any one of claims 1-9, wherein the therapeutically effective amount of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily, weekly, biweekly, once every four weeks or monthly.
11. The method of any one of claims 1-10, wherein the method further comprises administering a loading dose of the BCMA x CD3 bispecific antibody prior to the therapeutically effective amount of the BCMA x CD3 bispecific antibody.
12. The method of claim 11, wherein the loading dose comprises one or more step-up doses.
13. The method of claim 11 or 12, wherein the loading dose comprises 1, 2, 3, or more than 3 step up doses.
14. The method of claim 12 or 13, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are from about 60 pg/kg to about 6000 pg/kg.
15. The method of claim 14, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from 60 pg/kg, 240 pg/kg, or 300 pg/kg.
16. The method of claim 12 or 13, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are from about 3 mg to about 600 mg.
17. The method of claim 16, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg.
18. The method of claim 11 or 12 wherein the loading dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily every other day, or weekly.
19. The method of any one of claims 12 to 17, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are administered at a frequency selected from daily, every other day, or weekly.
20. The method of any one of claims 1-19, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
21. The method of any one of claims 1-20, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
22. The method of any one of claims 1-21, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
23. The method of any one of claims 1-22, wherein the anti-CD38 antibody is daratumumab.
24. The method of any one of claims 1-23, wherein the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg.
25. The method of any one of claims 1-24, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
26. The method of any one of claims 1-25, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from daily, weekly, biweekly, once every four weeks, or monthly.
27. The method of any one of claims 1-26, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
28. The method of any one of claims 1-27, wherein the IMiD is lenalidomide.
29. The method of any one of claims 1-28, wherein the therapeutically effective amount of the IMiD is from about 15 mg to about 50 mg.
30. The method of any one of claims 1-29, wherein the therapeutically effective amount of the IMiD is about 25 mg.
31. The method of any one of claims 1 -30, wherein the effective amount of the IMiD is administered at a frequency selected from daily or weekly.
32. The method of any one of claims 1-31, wherein the BCMA x CD3 bispecific antibody is administered subcutaneously.
33. The method of any one of claims 1-32, wherein the anti-CD38 antibody is administered subcutaneously.
34. The method of any one of claims 1-33, wherein the IMiD is administered orally.
35. The method of any one of claims 1-34, wherein the method further comprises administering a pre-treatment to the subject.
36. The method of claim 35, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
37. The method of claim 36, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
38. The method of claim 36 or 37, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
39. A method of treating a cancer in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of about 60 pg/kg, 240 pg/kg or 300 pg/kg or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg or 6000 pg/kg or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1200 mg to about 2400 mg; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 15 mg to about 50 mg; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
40. The method of claim 39, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
41. The method of claim 39 or 40, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
42. The method of any one of claims 39-41, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
43. The method of any one of claims 39-42, wherein the BCMA x CD3 bispecific antibody is teclistamab.
44. The method of any one of claims 39-43, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from about 60 pg/kg, 240 pg/kg, 300 pg/kg or any combination thereof.
45. The method of any one of claims 39-44, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are selected from about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof.
46. The method of any one of claims 39 to 45, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody are administered at a frequency selected from daily, every other day, or weekly.
47. The method of any one of claims 39 to 46, wherein the treatment dose of the BCMA x CD3 bispecific antibody is selected from 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof.
48. The method of any one of claims 39 to 46, wherein the treatment dose of the BCMA x CD3 bispecific antibody is selected from 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof.
49. The method of any one of claims 39 to 48, wherein the treatment dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
50. The method of any one of claims 39-49, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
51. The method of any one of claims 39-50, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
52. The method of any one of claims 39-51, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of
SEQ ID NO: 37.
53. The method of any one of claims 39-52, wherein the anti-CD38 antibody is daratumumab.
54. The method of any one of claims 39-53, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
55. The method of any one of claims 39-54, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
56. The method of any one of claims 39-55, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
57. The method of any one of claims 39-56, wherein the IMiD is lenalidomide.
58. The method of any one of claims 39-57, wherein the therapeutically effective amount of the IMiD is about 25 mg.
59. The method of any one of claims 39-58, wherein the therapeutically effective amount of the IMiD is administered at a frequency selected from daily or weekly.
60. The method of any one of claims 39-59, wherein the method further comprises administering a pre-treatment to the subject.
61. The method of claim 60, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
62. The method of claim 61, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
63. The method of claim 61 or 62, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
64. A method of treating a cancer in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody of 60 pg/kg, 240 pg/kg, or 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg, or any combination thereof at a frequency selected from, daily, every other day, or weekly; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, or 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, or 450 mg, or any combination thereof at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD) of about 25 mg at a frequency of daily or weekly; wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
65. The method of claim 64, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCM A binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO: 10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
66. The method of claim 64 or 65, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
67. The method of any one of claims 64-66, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
68. The method of any one of claims 64-67, wherein the BCMA x CD3 bispecific antibody is teclistamab.
69. The method of any one of claims 64-68, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
70. The method of any one of claims 64-69, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
71. The method of any one of claims 64-70, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
72. The method of any one of claims 64-71, wherein the anti-CD38 antibody is daratumumab.
73. The method of any one of claims 64-72, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
74. The method of any one of claims 64-73, wherein the IMiD is lenalidomide.
75. The method of any one of claims 64-74, wherein the method further comprises administering a pre-treatment to the subject.
76. The method of claim 75, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
77. The method of claim 76, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
78. The method of claim 76 or 77, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
79. A method of treating a cancer in a subject in need thereof, comprising: i) determining the subjects weight and assigning a BCMA x CD3 bispecific antibody treatment tier based on said weight, wherein if the subject’s weight is less than or equal to a predetermined threshold the subject is in tier 1, and wherein if the subject’s weight is greater than a pre-determined threshold the subject is in tier 2. ii) administering subcutaneously to the subject one or more step-up doses of a BCMA x CD3 bispecific antibody; iii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of the BCMA x CD3 bispecific antibody; iv) administering subcutaneously to the subject a therapeutically effective amount of an anti-CD38 antibody; and v) administering orally to the subject a therapeutically effective amount of an immunomodulatory drug (IMiD); wherein, the BCMA x CD3 bispecific antibody, the anti-CD38 antibody, and the IMiD are administered to the subject for a time sufficient to treat the cancer.
80. The method of claim 79, wherein the predetermined weight threshold is selected from the group consisting of 50 kg, 55 kg, 60 kg, 65 kg, or 70 kg.
81. The method of claim 79 or 80, wherein the predetermined weight threshold is 60 kg.
82. The method of any one of claims 79-81, wherein the BCMA x CD3 bispecific antibody comprises:
(1) a BCMA binding domain comprising a heavy chain variable region (VH) having heavy chain complementarity determining regions (HCDRs) HCDR1, HCDR2, and HCDR3, of the amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 8, respectively, and a light chain variable region (VL) having light chain complementarity determining regions (LCDRs) LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 9, SEQ ID NO:
10, and SEQ ID NO: 11, respectively, and
(2) a CD3 binding domain comprising a VH having HCDR1, HCDR2, and HCDR3 of the amino acid sequences of SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively, and a VL having LCDR1, LCDR2, and LCDR3 of the amino acid sequences of SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively.
83. The method of any one of claims 79-82, wherein the BCMA binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 18 and the VL having the amino acid sequence of SEQ ID NO: 19; and the CD3 binding domain comprises the VH having the amino acid sequence of SEQ ID NO: 20 and the VL having the amino acid sequence of SEQ ID NO: 21.
84. The method of any one of claims 79-83, wherein the BCMA x CD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 22, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 23, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 24, and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 25, wherein HC1 is associated with LC1 to form a first antigen -binding site that immunospecifically binds BCMA, and wherein HC2 is associated with LC2 to form a second antigen -binding site that immunoscpecifically binds CD3.
85. The method of any one of claims 79-84, wherein the BCMA x CD3 bispecific antibody is teclistamab.
86. The method of any one of claims 79-85, wherein the one or more step-up doses of a BCMA x CD3 bispecific antibody for a subject in tier 1 or tier 2 are selected from 3 mg, 4 mg, 15 mg, 24 mg, or 25 mg, or any combination thereof.
87. The method of claim 86, wherein the one or more step-up doses of a BCMA x CD3 bispecific antibody for a subject in tier 1 may or may not be different from the one or more step- up doses of a BCMA x CD3 bispecific antibody for a subject in tier 2.
88. The method of any one of claims 79-87, wherein the treatment dose of the BCMA x CD3 bispecific antibody for a subject in tier 1 or tier 2 are selected from 100 mg, 150 mg, 200 mg,
300 mg, or 450 mg, or any combination thereof.
89. The method of claim 88, wherein the treatment dose of a BCMA x CD3 bispecific antibody for a subject in tier 1 may or may not be different from the treatment dose of a BCMA x CD3 bispecific antibody for a subject in tier 2.
90. The method of any one of claims 79 to 89, wherein the one or more step-up doses of the BCMA x CD3 bispecific antibody is administered at a frequency selected from daily, every other day, or weekly.
91. The method of any one of claims 79 to 90, wherein the treatment dose of the BCMA x CD3 bispecific antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
92. The method of any one of claims 79-91, wherein the anti-CD38 antibody comprises a VH having an HCDR1, HCDR2, and HCDR3 or the amino acid sequences of SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively, and a VL having a LCDR1, LCDR2, and LCDR3, of the amino acid sequences of SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively.
93. The method of any one of claims 79-92, wherein the anti-CD38 antibody comprises the VH having the amino acid sequence of SEQ ID NO: 34, and the VL having the amino acid sequence of SEQ ID NO: 35.
94. The method of any one of claims 79-93, wherein the anti-CD38 antibody comprises a HC having the amino acid sequence of SEQ ID NO: 36, and a LC having the amino acid sequence of SEQ ID NO: 37.
95. The method of any one of claims 79-94, wherein the anti-CD38 antibody is daratumumab.
96. The method of any one of claims 79-95, wherein the therapeutically effective amount of the anti-CD38 antibody is from about 1200 mg to about 2400 mg.
97. The method of any one of claims 79-96, wherein the therapeutically effective amount of the anti-CD38 antibody is about 1800 mg.
98. The method of any one of claims 79-97, wherein the therapeutically effective amount of the anti-CD38 antibody is administered at a frequency selected from weekly, biweekly, once every four weeks, or monthly.
99. The method of any one of claims 79-98, wherein the IMiD is selected from the group consisting of thalidomide, pomalidomide, lenalidomide, or any combination thereof.
100. The method of any one of claims 79-99, wherein the IMiD is lenalidomide.
101. The method of any one of claims 79-100, wherein the therapeutically effective amount of the IMiD is from about 15 mg to about 50 mg.
102. The method of any one of claims 79-101, wherein the therapeutically effective amount of the IMiD is about 25 mg.
103. The method of any one of claims 79-102, wherein the effective amount of the IMiD is administered at a frequency selected from daily or weekly.
104. The method of any one of claims 79-103, wherein the method further comprises administering a pre-treatment to the subject.
105. The method of claim 104, wherein the pretreatment regimen comprises administering 8 mg to 40 mg of dexamethasone, 25 mg to 50 mg of diphenhydramine, and 650 mg to 1000 mg acetaminophen.
106. The method of claim 105, wherein: i) the dexamethasone is administered on Days 1, 2, 4, and 8 of a first treatment cycle, and weekly thereafter for a second, third, and fourth treatment cycles; ii) the diphenhydramine is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody; and iii) the acetaminophen is administered for all doses of the anti-CD38 antibody and all step-up and first treatment dose of the BCMA x CD3 bispecific antibody.
107. The method of claim 105 or 106, wherein the pre-treatment regimen further comprises montelukast administered at 10 mg.
108. The method of any one of claims 1-107, wherein the BCMA x CD3 bispecific antibody, anti-CD38 antibody, and IMiD are administered on a 28-day treatment cycle.
109. The method of claim 108, wherein the method further comprises treating the subject for 1, 2, 3, 4, 5, 6, 7, or more than 7 treatment cycles.
110. The method of claim 108 or 109, wherein the frequency, dose, or a combination thereof of the BCMA x CD3 bispecific antibody, anti-CD38 antibody, IMiD, or combination thereof may or may not be changed between a current and a subsequent treatment cycle.
111. The method of any one of claims 1 to 110, wherein the anti-CD38 antibody is administered or provided for administration together with a hyaluronidase.
112. The method of claim 111, wherein the hyaluronidase is rHuPH20.
113. The method of claim 111 or 112, wherein the hyaluronidase is administered or provided at 30,000 U.
114. The method of any one of claims 1-113, wherein the cancer is multiple myeloma.
115. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject one or more step-up doses of teclistamab at 60 pg/kg, 240 pg/kg, 300 pg/kg, or any combination thereof, or of about 3 mg, 4 mg, 15 mg, 24 mg, 25 mg, or any combination thereof, on days 2 and 4 of the start of treatment; ii) after the one or more step-up doses, administering subcutaneously to the subject a treatment dose of teclistamab of about 720 pg/kg, 1500 pg/kg, 3000 pg/kg, 6000 pg/kg, or any combination thereof, or of about 100 mg, 150 mg, 200 mg, 300 mg, 450 mg or any combination thereof, at a frequency selected from weekly, biweekly, once every four weeks, or once a month; iii) administering subcutaneously to the subject a therapeutically effective amount of dartumumab of about 1800 mg at a frequency selected from weekly, biweekly, once every four weeks, or once a month; and iv) administering orally to the subject a therapeutically effective amount of lenalidomide of about 25 mg at a frequency of once per day; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
116. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab of 60 pg/kg, on day 2 of a treatment regimen, and administering subcutaneously to the subject a second step-up dose of 240 pg/kg or 300 pg/kg on day 4 of a treatment regimen; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab of
720 pg/kg or 1500 pg/kg, on days 8, 15, and 22 of the first treatment cycle; b) administering subcutaneously to the subject a treatment dose of teclistamab of
720 pg/kg or 1500 pg/kg weekly for each treatment cycle thereafter; wherein the 720 g/kg weekly dose optionally is elevated to 1500 g/kg weekly; wherein the 1500 pg/kg weekly dose optionally is elevated to 3000 pg/kg biweekly starting at a third treatment cycle; and wherein the 3000 pg/kg biweekly dose optionally is elevated to 6000 pg/kg once every four weeks starting at a seventh treatment cycle; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
117. A method of treating multiple myeloma in a subject in need thereof, comprising: i) administering subcutaneously to the subject a first step-up dose of teclistamab on day 2 of a first treatment cycle and a second step-up dose of teclistamab on day 4 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the first step-up dose is 3 mg teclistamab and the second step-up dose is 15 mg teclistamab, wherein if the subject weight greater than 60 kg then the first step-up dose is 4 mg teclistamab and the second step-up dose is 24 mg or 25 mg teclistamab; ii) after the first and second step-up doses, administering a treatment dose of teclistamab based on a 28 day treatment cycle comprising, a) administering subcutaneously to the subject a treatment dose of teclistamab, on days 8, 15, and 22 of the first treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; b) administering subcutaneously to the subject a treatment dose of teclistamab, weekly for a second treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 100 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is 150 mg; c) administering subcutaneously to the subject a treatment dose of teclistamab, biweekly for a third, fourth, fifth, and sixth treatment cycle, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, wherein if the subject weighs greater than 60 kg then the treatment dose is
300 mg; and d) administering subcutaneously to the subject a treatment dose of teclistamab, once every four weeks for a seventh and subsequent treatment cycles, wherein if the subject weighs less than or equal to 60 kg then the treatment dose is 200 mg, optionally wherein the treatment dose is elevated to 300 mg, and wherein if the subject weighs greater than 60 kg then the treatment dose is 300 mg, optionally wherein the treatment dose is elevated to 450 mg; iii) administering subcutaneously to the subject 1800 mg dartumumab on a 28 day treatment cycle, wherein the daratumumab is administered weekly for the first and second treatment cycle, biweekly for the third through sixth treatment cycle, and once every four weeks for the seventh and subsequent treatment cycles; and iv) administering orally to the subject 25mg lenalidomide at a frequency of once per day for 21 days of a 28 day treatment cycle, wherein the lenalidomide treatment starts at treatment cycle 2; wherein, teclistamab, daratumumab, and lenalidomide are administered to the subject for a time sufficient to treat the cancer.
118. The method of any one of claims 115-117, wherein the method further comprises administering a pre-treatment regimen.
119. The method of claim 118, wherein the pre-treatment regimen comprises: i) intravenously or orally administering a therapeutically effective amount of dexamethasone, wherein the dexamethasone is administered at 20 mg on Day 1 of a first treatment cycle, 16 mg on days 2, 4, 8, 15, and 22 of a first treatment cycle, and 20 mg or 40 mg weekly during each of a second, third, and fourth treatment cycles; ii) intravenously or orally administering a therapeutically effective amount of diphenhydramine, wherein the diphenhydramine is administered at 25 mg to 50 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab; and iii) intravenously or orally administering a therapeutically effective amount of acetaminophen, wherein the acetaminophen is administered at 650 mg to 1000 mg for all doses of daratumumab and all step-up doses and the first treatment dose of teclistamab.
120. The method of claim 119, wherein the diphenhydramine and acetaminophen are further administered after any Grade 2 or greater cytokine release syndrome or administration related reaction to the teclistamab.
121. The method of claim 119 or 120, wherein the dexamethasone is further administered after any Grade 2, 3, or greater than 3 administration related reaction to the daratumumab, and/or any Grade 2 or greater cytokine release syndrome or administration related reaction to the teclistamab.
122. The method of any one of claims 118-121, wherein the pre-treatment regimen further comprises administration of montelukast at 10 mg.
PCT/IB2023/060998 2022-11-02 2023-11-01 Methods of treating cancers WO2024095173A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263382089P 2022-11-02 2022-11-02
US63/382,089 2022-11-02

Publications (1)

Publication Number Publication Date
WO2024095173A1 true WO2024095173A1 (en) 2024-05-10

Family

ID=88731546

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/060998 WO2024095173A1 (en) 2022-11-02 2023-11-01 Methods of treating cancers

Country Status (1)

Country Link
WO (1) WO2024095173A1 (en)

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988001649A1 (en) 1986-09-02 1988-03-10 Genex Corporation Single polypeptide chain binding molecules
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
WO1994013804A1 (en) 1992-12-04 1994-06-23 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
WO1998044001A1 (en) 1997-03-27 1998-10-08 Commonwealth Scientific And Industrial Research Organisation High avidity polyvalent and polyspecific reagents
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
WO2004078140A2 (en) 2003-03-05 2004-09-16 Halozyme, Inc. SOLUBLE HYALURONIDASE GLYCOPROTEIN (sHASEGP), PROCESS FOR PREPARING THE SAME, USES AND PHARMACEUTICAL COMPOSITIONS COMPRISING THEREOF
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
US20070287170A1 (en) 2006-03-24 2007-12-13 Merck Patent Gmbh Engineered heterodimeric protein domains
WO2007147901A1 (en) 2006-06-22 2007-12-27 Novo Nordisk A/S Production of bispecific antibodies
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
WO2009134776A2 (en) 2008-04-29 2009-11-05 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
US20100015133A1 (en) 2005-03-31 2010-01-21 Chugai Seiyaku Kabushiki Kaisha Methods for Producing Polypeptides by Regulating Polypeptide Association
US20100028637A1 (en) 2005-06-22 2010-02-04 Sunjuet Deutschland Gmbh Multi-Layer Film Comprising a Barrier Layer and an Antistatic Layer
US7829673B2 (en) 2005-03-23 2010-11-09 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
US20110123532A1 (en) 2009-04-27 2011-05-26 Oncomed Pharmaceuticals, Inc. Method for Making Heteromultimeric Molecules
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
WO2011143545A1 (en) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Heterodimeric proteins and methods for producing and purifying them
US8088896B2 (en) 2005-10-12 2012-01-03 Morphosys Ag Generation and profiling of fully human gold-derived therapeutic antibodies specific for human CD38
WO2012022811A1 (en) 2010-08-20 2012-02-23 Leadartis, S.L. Engineering multifunctional and multivalent molecules with collagen xv trimerization domain
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
US20120149876A1 (en) 2010-11-05 2012-06-14 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
WO2013096291A2 (en) 2011-12-20 2013-06-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
US20130195849A1 (en) 2011-11-04 2013-08-01 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
WO2013157954A1 (en) 2012-04-20 2013-10-24 Merus B.V. Methods and means for the production of ig-like molecules
US20140303356A1 (en) 2011-10-27 2014-10-09 Michael Gramer Production of heterodimeric proteins
WO2017031104A1 (en) 2015-08-17 2017-02-23 Janssen Pharmaceutica Nv Anti-bcma antibodies, bispecific antigen binding molecules that bind bcma and cd3, and uses thereof
US20180118849A1 (en) 2016-09-30 2018-05-03 Hoffmann-La Roche Inc. Bispecific t cell activating antigen binding molecules
WO2021228783A1 (en) * 2020-05-11 2021-11-18 Janssen Pharmaceutica Nv Methods for treating multiple myeloma
WO2021229507A2 (en) * 2020-05-13 2021-11-18 Pfizer Inc. Methods, therapies and uses for treating cancer

Patent Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988001649A1 (en) 1986-09-02 1988-03-10 Genex Corporation Single polypeptide chain binding molecules
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5403484A (en) 1988-09-02 1995-04-04 Protein Engineering Corporation Viruses expressing chimeric binding proteins
US5571698A (en) 1988-09-02 1996-11-05 Protein Engineering Corporation Directed evolution of novel binding proteins
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5580717A (en) 1990-05-01 1996-12-03 Affymax Technologies N.V. Recombinant library screening methods
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5969108A (en) 1990-07-10 1999-10-19 Medical Research Council Methods for producing members of specific binding pairs
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
US6593081B1 (en) 1991-12-02 2003-07-15 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US6582915B1 (en) 1991-12-02 2003-06-24 Medical Research Council Production of anti-self bodies from antibody segment repertories and displayed on phage
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US6555313B1 (en) 1991-12-02 2003-04-29 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US6521404B1 (en) 1991-12-02 2003-02-18 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US6544731B1 (en) 1991-12-02 2003-04-08 Medical Research Council Production of anti-self antibodies from antibody segment repertories and displayed on phage
WO1994013804A1 (en) 1992-12-04 1994-06-23 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
WO1998044001A1 (en) 1997-03-27 1998-10-08 Commonwealth Scientific And Industrial Research Organisation High avidity polyvalent and polyspecific reagents
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
WO2004078140A2 (en) 2003-03-05 2004-09-16 Halozyme, Inc. SOLUBLE HYALURONIDASE GLYCOPROTEIN (sHASEGP), PROCESS FOR PREPARING THE SAME, USES AND PHARMACEUTICAL COMPOSITIONS COMPRISING THEREOF
US7829673B2 (en) 2005-03-23 2010-11-09 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
US20100015133A1 (en) 2005-03-31 2010-01-21 Chugai Seiyaku Kabushiki Kaisha Methods for Producing Polypeptides by Regulating Polypeptide Association
US20100028637A1 (en) 2005-06-22 2010-02-04 Sunjuet Deutschland Gmbh Multi-Layer Film Comprising a Barrier Layer and an Antistatic Layer
US8088896B2 (en) 2005-10-12 2012-01-03 Morphosys Ag Generation and profiling of fully human gold-derived therapeutic antibodies specific for human CD38
US20070287170A1 (en) 2006-03-24 2007-12-13 Merck Patent Gmbh Engineered heterodimeric protein domains
WO2007147901A1 (en) 2006-06-22 2007-12-27 Novo Nordisk A/S Production of bispecific antibodies
US20090182127A1 (en) 2006-06-22 2009-07-16 Novo Nordisk A/S Production of Bispecific Antibodies
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
WO2009134776A2 (en) 2008-04-29 2009-11-05 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
US20110123532A1 (en) 2009-04-27 2011-05-26 Oncomed Pharmaceuticals, Inc. Method for Making Heteromultimeric Molecules
US9150663B2 (en) 2010-04-20 2015-10-06 Genmab A/S Heterodimeric antibody Fc-containing proteins and methods for production thereof
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
WO2011143545A1 (en) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Heterodimeric proteins and methods for producing and purifying them
WO2012022811A1 (en) 2010-08-20 2012-02-23 Leadartis, S.L. Engineering multifunctional and multivalent molecules with collagen xv trimerization domain
US20120149876A1 (en) 2010-11-05 2012-06-14 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
US20140303356A1 (en) 2011-10-27 2014-10-09 Michael Gramer Production of heterodimeric proteins
US20130195849A1 (en) 2011-11-04 2013-08-01 Zymeworks Inc. Stable Heterodimeric Antibody Design with Mutations in the Fc Domain
WO2013096291A2 (en) 2011-12-20 2013-06-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
WO2013157954A1 (en) 2012-04-20 2013-10-24 Merus B.V. Methods and means for the production of ig-like molecules
WO2017031104A1 (en) 2015-08-17 2017-02-23 Janssen Pharmaceutica Nv Anti-bcma antibodies, bispecific antigen binding molecules that bind bcma and cd3, and uses thereof
US20180118849A1 (en) 2016-09-30 2018-05-03 Hoffmann-La Roche Inc. Bispecific t cell activating antigen binding molecules
WO2021228783A1 (en) * 2020-05-11 2021-11-18 Janssen Pharmaceutica Nv Methods for treating multiple myeloma
WO2021229507A2 (en) * 2020-05-13 2021-11-18 Pfizer Inc. Methods, therapies and uses for treating cancer

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
ADRIOUCH ET AL., MICROBES INFECT, vol. 14, 2012, pages 1284 - 92
ANSELL S M ET AL: "Denileukin diftitox in combination with rituximab for previously untreated follicular B-cell non-Hodgkin's lymphoma", BLOOD CANCER JOURNAL, NATURE PUBLISHING GROUP UK, LONDON, vol. 26, no. 5, 21 October 2011 (2011-10-21), pages 1046 - 1052, XP037785278, ISSN: 0887-6924, [retrieved on 20111021], DOI: 10.1038/LEU.2011.297 *
BAERT ET AL., N ENGL J MED, vol. 348, 2003, pages 602 - 08
CHAMESBATY, CURR OPIN DRUG DISC DEV, vol. 12, 2009, pages 276
CHIARUGI ET AL., NATURE REVIEWS, vol. 12, 2012, pages 741 - 52
CHOTHIA ET AL., J MOL BIOL, vol. 196, 1987, pages 901 - 17
FUNARO ET AL., J IMMUNOL, vol. 145, 1990, pages 2390 - 6
GUSE ET AL., NATURE, vol. 398, 1999, pages 70 - 3
HONEGGERPLUCKTHUN, J MOL BIOL, vol. 309, 2001, pages 657 - 70
HOOGENBOOMWINTER, J MOL BIOL, vol. 222, 1991, pages 581
KNAPPIK ET AL., J MOL BIOL, vol. 296, 2000, pages 57 - 86
KREBS ET AL., J IMMUNOL METH, vol. 254, 2001, pages 67 - 84
LEFRANC ET AL., DEV COMP IMMUNOL, vol. 27, 2003, pages 55 - 77
MARTINTHORNTON, J BMOL BIOL, vol. 263, 1996, pages 800 - 15
MOORE DONALD C. ET AL: "Evaluation of Montelukast for the Prevention of Infusion-related Reactions With Daratumumab", CLINICAL LYMPHOMA MYELOMA AND LEUKEMIA, vol. 20, no. 10, 7 June 2020 (2020-06-07), NL, pages e777 - e781, XP093123126, ISSN: 2152-2650, DOI: 10.1016/j.clml.2020.05.024 *
NUNEZ-PRADO ET AL., DRUG DISCOVERY TODAY, vol. 20, no. 5, 2015, pages 588 - 594
SHEETS ET AL., PITAS (USA, vol. 95, 1998, pages 6157 - 62
SHI ET AL., J MOL BIOL, vol. 397, 2010, pages 385 - 96
SHI ET AL., J. MOL. BIOL., vol. 397, 2010, pages 385 - 96
STICKLER ET AL., GENES AND IMMUNITY, vol. 12, 2011, pages 213 - 21
TERHORST ET AL., CELL, 1981, pages 771 - 80
VAUGHAN ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 309 - 14
WU ET AL., J EXP MED, vol. 132, 1970, pages 211 - 50

Similar Documents

Publication Publication Date Title
US20240101697A1 (en) Methods of treating cancers and enhancing efficacy of t cell redirecting therapeutics
US20230272102A1 (en) Methods of treating cancers and enhancing efficacy of bcmaxcd3 bispecific antibodies
KR20230009450A (en) Methods for Treating Multiple Myeloma
IL294017A (en) Materials and methods for in vivo biological targeting
US20220177584A1 (en) Methods for treating multiple myeloma
WO2024095173A1 (en) Methods of treating cancers
US20230295292A1 (en) Methods of treating cancers and enhancing efficacy of gprc5dxcd3 bispecific antibodies
US20210363252A1 (en) Compositions comprising a t cell redirection therapeutic and a vla-4 adhesion pathway inhibitor
Segués Cisteró Antibody-based approaches to modulate immune response
EA045878B1 (en) BISPECIFIC ANTIBODIES TO BCMA/CD3 AND GPRDC5D/CD3 FOR USE IN CANCER TREATMENT