WO2024069240A2 - Thérapie de combinaison de protéines de fusion se liant à cd38 - Google Patents

Thérapie de combinaison de protéines de fusion se liant à cd38 Download PDF

Info

Publication number
WO2024069240A2
WO2024069240A2 PCT/IB2023/000653 IB2023000653W WO2024069240A2 WO 2024069240 A2 WO2024069240 A2 WO 2024069240A2 IB 2023000653 W IB2023000653 W IB 2023000653W WO 2024069240 A2 WO2024069240 A2 WO 2024069240A2
Authority
WO
WIPO (PCT)
Prior art keywords
fusion protein
subject
binding fusion
administered
seq
Prior art date
Application number
PCT/IB2023/000653
Other languages
English (en)
Other versions
WO2024069240A3 (fr
Inventor
Sabrina Collins
Xavier PAROT
Shuang Wu LI
Laura THESILLAT-VERSMEE
Richard LABOTKA
Jim Sampson
Neeraj Gupta
Shining WANG
Kaveri SURYANARAYAN
Original Assignee
Takeda Pharmaceutical Company Limited
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 Takeda Pharmaceutical Company Limited filed Critical Takeda Pharmaceutical Company Limited
Publication of WO2024069240A2 publication Critical patent/WO2024069240A2/fr
Publication of WO2024069240A3 publication Critical patent/WO2024069240A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2497Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing N- glycosyl compounds (3.2.2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/02Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2) hydrolysing N-glycosyl compounds (3.2.2)
    • C12Y302/02005NAD+ nucleosidase (3.2.2.5)

Definitions

  • CD38 is a 45 kDa type II transmembrane glycoprotein. It has a short N-terminal cytoplasmic tail of 20 amino acids, a single transmembrane helix and a long extracellular domain of 256 amino acids. It is expressed on the surface of many immune cells including CD4 and CD8 positive T cells, B cells, NK cells, monocytes, plasma cells, and on a significant proportion of normal bone marrow precursor cells.
  • CD38 is expressed at high levels on various types of cancer cells, e.g., multiple myeloma cells, in most cases of T- and B-lineage acute lymphoblastic leukemias, some acute myelocytic leukemias, follicular center cell lymphomas and T lymphoblastic lymphomas. CD38 is also expressed on B-lineage chronic lymphoblastic leukemia (B-CLL) cells. Antibodies that target CD38 have been used in the treatment of CD38-expressing cancers and hematological malignancies.
  • B-CLL B-lineage chronic lymphoblastic leukemia
  • Interferons and in particular IFN-alpha, are able to increase apoptosis and decrease proliferation of certain cancer cells.
  • IFN-alpha has been approved by the FDA for the treatment of several cancers including melanoma, renal cell carcinoma, B cell lymphoma, multiple myeloma, chronic myelogenous leukemia (CML) and hairy cell leukemia.
  • IFN may be targeted to cancer cells, for example, by linking it with a targeting antibody or targeting fragment thereof.
  • the present disclosure in some aspects, relates to methods of treating cancer (e.g.,CD38-expressing cancer) using a CD38-binding fusion protein comprising an anti-CD38 binder, such as an-anti CD38 antibody or binding domain or fragment thereof, fused to one or more (e.g., one, two) attenuated interferon alpha-2b protein in combination with one or more (e.g., one, two, or more) agents for treating the cancer.
  • the one or more agents comprise an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide) and/or a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • the CD38-expressing cancer is multiple myeloma.
  • the multiple myeloma is Relapsed or Refractory Multiple Myeloma (RRMM).
  • the methods described herein are effective in treating RRMM: (i) in subjects who have received prior lines of therapy (e.g., one or more prior lines of therapy) for multiple myeloma, including at least one proteasome inhibitor, at least one anti-CD38 monoclonal antibody (mAb) drug, and/or at least one immunomodulatory drug (e.g., an immunomodulatory imide drug (IMiD)); (ii) in subjects who were refractory to a proteasome inhibitor, an immunomodulatory drug (e.g., an IMiD (e.g., lenalidomide or pomalidomide)), or an anti-CD38 antibody therapy regardless of prior lines of therapy; and/or (iii) in subjects who demonstrated disease progression with one or more prior lines of therapy.
  • prior lines of therapy e.g.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein, an immunomodulatory drug, and a proteasome inhibitor, wherein the CD38- binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha- 2b.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b, and wherein the subject is receiving or has received treatment with an immunomodulatory drug, and a proteasome inhibitor.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof an immunomodulatory drug, and a proteasome inhibitor, wherein the subject is receiving or has received treatment with a CD38-binding fusion protein, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein and an immunomodulatory drug, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b, and wherein the subject is receiving or has received treatment with a proteasome inhibitor.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a proteasome inhibitor, wherein the subject is receiving or has received treatment with a CD38-binding fusion protein and an immunomodulatory drug, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein and a proteasome inhibitor, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to an attenuated interferon alpha-2b, and wherein the subject is receiving or has received treatment with and one or more immunomodulatory drug.
  • Some aspects of the present disclosure provide methods of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof an immunomodulatory drug, wherein the subject is receiving or has received treatment with a CD38-binding fusion protein and a proteasome inhibitor, and wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b.
  • the proteasome inhibitor is bortezomib or carfilzomib.
  • the immunomodulatory drug is lenalidomide or pomalidomide.
  • the anti-CD38 antibody comprises a heavy chain complementarity determining region 1 (CDR-H1) comprising the amino acid sequence of SEQ ID NO: 1, a heavy chain complementarity determining region 2 (CDR-H2) comprising the amino acid sequence of SEQ ID NO: 2, a heavy chain complementarity determining region 3 (CDR-H3) comprising the amino acid sequence of SEQ ID NO: 3, a light chain complementarity determining region 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO: 4, a light chain complementarity determining region 2 (CDR-L2) comprising the amino acid sequence of SEQ ID NO: 5, and a light chain complementarity determining region 3 (CDR-L3) comprising the amino acid sequence of SEQ ID NO: 6.
  • CDR-H1 comprising the amino acid sequence of SEQ ID NO: 1
  • CDR-H2 comprising the amino acid sequence of SEQ ID NO: 2
  • CDR-H3 heavy chain complementarity determining region 3
  • the anti-CD38 antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 8.
  • the anti-CD38 antibody is a full-length IgG antibody.
  • the anti-CD38 antibody comprises a human IgG4 constant region.
  • the human IgG4 constant region comprises a proline at position 228 according to the EU numbering system.
  • the anti-CD38 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 9 and a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • the attenuated interferon alpha-2b comprises T106A and A145D mutations relative to an interferon alpha- 2b comprising the amino acid sequence of SEQ ID NO: 11. In some embodiments, the attenuated interferon alpha- 2b comprises the amino acid sequence of SEQ ID NO: 12. In some embodiments, the attenuated interferon alpha-2b is fused to the C-terminus of the heavy chain. In some embodiments, the CD38-binding fusion protein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 13 and a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • the CD38-binding fusion protein is administered once every 4 weeks during a period of administration. In some embodiments, the administrations are according to a 4-week treatment cycle during a period of administration. In some embodiments, the period of administration is up to 5 years.
  • the subject is administered 50-250 mg of the CD38-binding fusion protein each 4-week treatment cycle. In some embodiments, the subject is administered 60 mg, 80 mg, 120 mg, or 240 mg of the CD38-binding fusion protein each 4-week treatment cycle. In some embodiments, the subject is administered the CD38-binding fusion protein at a dose of 0.5-4 mg/kg each 4-week treatment cycle. In some embodiments, the subject is administered the CD38-binding fusion protein at a dose of 0.75 mg/kg, 1 mg/kg, 2 mg/kg, or 3 mg/kg each 4-week treatment cycle. In some embodiments, administration of the CD38- binding fusion protein occurs on day 1 of each 4-week treatment cycle. In some embodiments, the CD38-binding fusion protein is administered intravenously.
  • the immunomodulatory drug is pomalidomide.
  • the subject is administered 4 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle during the period of administration.
  • pomalidomide is administered orally.
  • the immunomodulatory drug is lenalidomide.
  • the subject is administered 10 mg of lenalidomide daily for 28 days of each 4- week treatment cycle during the period of administration.
  • lenalidomide is administered orally.
  • the proteasome inhibitor is bortezomib.
  • bortezomib is administered at a dose of 1.3 mg/m 2 on days 1, 8, 15 for 8 4-week treatment cycles and on days 1 and 8 for remaining 4-week treatment cycles during the period of administration.
  • bortezomib is administered subcutaneously.
  • the proteasome inhibitor is carfilzomib.
  • carfilzomib is administered at a dose of 20 mg/m 2 on day 1 of the first four- week treatment cycle, and at a dose of up to 70 mg/m 2 on days 8 and 15 for remaining 4-week treatment cycles during the period of administration.
  • carfilzomib is administered intravenously.
  • the CD38-binding fusion protein is in a composition that further comprises histidine, arginine, sucrose, and polysorbate 80 (PS80).
  • the composition comprises, histidine at a concentration of 50 nM, arginine at a concentration of 100 nM, sucrose at a concentration of 50 mg/ml, and PS80 at a concentration of 0.2 mg/ml.
  • the composition is at a pH of 6.6.
  • the subject is receiving or has received dexamethasone.
  • the CD38-expressing cancer is multiple myeloma. In some embodiments, multiple myeloma is refractory multiple myeloma. In some embodiments, the multiple myeloma is previously untreated multiple myeloma. In some embodiments, the administration results in tumor regression.
  • the subject is human. In some embodiments, the subject is a rodent.
  • CD38-binding fusion proteins for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof, an immunomodulatory drug, and a proteasome inhibitor, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha- 2b.
  • CD38-binding fusion proteins for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b, and wherein the subject is receiving or has received treatment with an immunomodulatory drug and a proteasome inhibitor.
  • Some aspects of the present disclosure provide lenalidomide or pomalidomide, and a proteasome inhibitor for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof an immunomodulatory drug and a proteasome inhibitor, wherein the subject is receiving or has received treatment with a CD38- binding fusion protein, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha- 2b.
  • Some aspects of the present disclosure provide CD38-binding fusion proteins and lenalidomide or pomalidomide for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein and an immunomodulatory drug, wherein the CD38-binding fusion protein comprises an anti- CD38 antibody fused to one or more attenuated interferon alpha-2b, and wherein the subject is receiving or has received treatment with a proteasome inhibitor.
  • Some aspects of the present disclosure provide proteasome inhibitor for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a proteasome inhibitor, wherein the subject is receiving or has received treatment with a CD38-binding fusion protein and an immunomodulatory drug, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha- 2b.
  • Some aspects of the present disclosure provide CD38-binding fusion proteins and a proteasome inhibitor for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof a CD38-binding fusion protein and a proteasome inhibitor, wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha- 2b, and wherein the subject is receiving or has received treatment with and an immunomodulatory drug.
  • Some aspects of the present disclosure provide an immunomodulatory drug for use in a method of treating a CD38-expressing cancer, the method comprising administering to a subject in need thereof the immunomodulatory drug, wherein the subject is receiving or has received treatment with a CD38-binding fusion protein and a proteasome inhibitor, and wherein the CD38-binding fusion protein comprises an anti-CD38 antibody fused to one or more attenuated interferon alpha-2b.
  • the proteasome inhibitor is bortezomib or carfilzomib.
  • the immunomodulatory drug is lenalidomide or pomalidomide.
  • Some aspects of the present disclosure provide a method of treating a subject having multiple myeloma, the method comprising administering to the subject: (i) 60-240 mg of CD38-binding fusion protein once during a 4-week treatment cycle; and (ii) 2-4 mg of pomalidomide daily for up to the first 21 days of the 4-week treatment cycle.
  • 2 mg of pomalidomide is administered to the subject.
  • 3 mg of pomalidomide is administered to the subject.
  • 4 mg of pomalidomide is administered to the subject.
  • Some aspects of the present disclosure provide a method of treating a subject having multiple myeloma, the method comprising administering to the subject: (i) 60-240 mg of CD38-binding fusion protein once during a 4-week treatment cycle; and (ii) 10 mg of lenalidomide daily for up to 28 days during the 4-week treatment cycle.
  • 15 mg lenalidomide is administered to the subject after the 4- week treatment cycle.
  • Some aspects of the present disclosure provide a method of treating a subject having multiple myeloma, the method comprising administering to the subject: (i) 60-240 mg of CD38-binding fusion protein once during a 4-week treatment cycle; and (ii) 1.3 mg/m2 of bortezomib on days 8, 15 and 22 for up to the first eight 4-week treatment cycles.
  • 1.3 mg/m2 of bortezomib is administered on days 8 and 22 at least one 4-week treatment cycle that occurs after the first eight 4-week treatment cycles.
  • 60 mg of the CD38-binding fusion protein is administered. In some embodiments, 80 mg of the CD38-binding fusion protein is administered. In some embodiments, 120 mg of the CD38-binding fusion protein is administered. In some embodiments, 240 mg of the CD38-binding fusion protein is administered.
  • the CD38-binding fusion protein comprises: (i) a first anti-CD38 antibody comprising a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 8; and (ii) an attenuated interferon alpha-2b comprising an amino acid sequence of SEQ ID NO: 12.
  • FIGs. 1A-1B show the tumor inhibition activity of CD38-binding fusion protein combined with lenalidomide (FIG.1A) and bortezomib (FIG. IB) in NCI-H929 multiple myeloma (MM) xenograft tumor model.
  • mice bearing subcutaneous NCI-H929 tumors were randomized and treated with vehicle (PBS, twice a week (BIW) for 3 weeks), a CD38-binding fusion protein (amino acid sequences shown in Table 1, 2.5 mg/kg, BIW for 3 weeks), lenalidomide (25 mg/kg, BIW for 3 weeks), or bortezomib (1.0 mg/kg, BIW for 3 weeks), or the combination of a CD38-binding fusion protein and lenalidomide, or the combination of a CD38-binding fusion protein and bortezomib (at the same dosing amounts and schedules as the single doses).
  • Individual tumor volumes were measured BIW and the graphs show median tumor volume per treatment arm.
  • the present disclosure in some aspects, relates to methods of treating cancer (e.g., CD38-expressing cancer) using a CD38-binding fusion protein comprising an anti-CD38 antibody fused to one or more (e.g., one, two) attenuated interferon alpha- 2b protein in combination with one or more (e.g., one, two, or more) agents for treating the cancer.
  • the one or more agents comprise an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide) and/or a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • the CD38-expressing cancer is multiple myeloma.
  • the multiple myeloma is relapsed or refractory multiple myeloma.
  • the methods described herein are effective in treating relapsed or refractory multiple myeloma: (i) in subjects who have received prior lines of therapy (e.g., one or more prior lines of therapy) for multiple myeloma, including at least one proteasome inhibitor, at least one anti-CD38 monoclonal antibody (mAb) drug, and/or at least one immunomodulatory drug (e.g., an IMiD); (ii) in subject who were refractory to a proteasome inhibitor, an immunomodulatory drug (e.g., an ImiD), or an anti-CD38 antibody therapy regardless of prior lines of therapy; and/or (iii) in subjects who demonstrated disease progression with one or more prior lines of therapy.
  • prior lines of therapy e.g., one or more prior lines of therapy
  • mAb mono
  • a CD38-binding fusion protein comprising an anti- CD38 antibody fused to one or more (e.g., one, two) attenuated interferon alpha- 2b protein in combination with an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide) and a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a proteasome inhibitor e.g., bortezomib or carfilzomib.
  • the subject being treated in a method described herein is not also receiving treatments with an anti-CD38 antibody (e.g., daratumumab).
  • the subject being treated in a method described herein may have previously received treatments with an anti- CD38 antibody (e.g., daratumumab), and maybe refractory
  • a method described herein comprises administering to a subject in need thereof a CD38-binding fusion protein described herein, an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide), and a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a proteasome inhibitor e.g., bortezomib or carfilzomib.
  • a method described herein comprises administering to a subject in need thereof a CD38-binding fusion protein described herein, pomalidomide, and carfilzomib.
  • a method described herein comprises administering to a subject in need thereof a CD38-binding fusion protein described herein, wherein the subject is receiving or has received treatment with an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide) and a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a proteasome inhibitor e.g., bortezomib or carfilzomib
  • a method described herein comprises administering to a subject in need thereof an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide) and a proteasome inhibitor (e.g., bortezomib or carfilzomib), wherein the subject is receiving or has received treatment with a CD38-binding fusion protein described herein.
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a proteasome inhibitor e.g., bortezomib or carfilzomib
  • a method described herein comprises administering to a subject in need thereof a CD38-binding fusion protein described herein and an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide), wherein the subject is receiving or has received treatment with a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a method described herein comprises administering to a subject in need thereof a proteasome inhibitor (e.g., bortezomib or carfilzomib), wherein the subject is receiving or has received treatment with a CD38-binding fusion protein described herein and an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide).
  • a proteasome inhibitor e.g., bortezomib or carfilzomib
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a method described herein comprises administering to a subject in need thereof a CD38-binding fusion protein described herein and a proteasome inhibitor (e.g., bortezomib or carfilzomib), wherein the subject is receiving or has received treatment with an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide).
  • a proteasome inhibitor e.g., bortezomib or carfilzomib
  • an immunomodulatory drug e.g., an IMiD such as lenalidomide or pomalidomide
  • a method described herein comprises administering to a subject in need thereof an immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide), wherein the subject is receiving or has received treatment with a CD38-binding fusion protein described herein and a proteasome inhibitor (e.g., bortezomib or carfilzomib).
  • a CD38-binding fusion protein refers to a fusion protein comprising a CD38 binder, such as an anti-CD38 antibody or binding domain or fragment thereof fused to one or more (e.g., one, two) attenuated interferon alpha-2b proteins.
  • a “fusion protein” refers to a polypeptide comprising two or more proteinaceous components associated by at least one covalent bond which is a peptide bond, regardless of whether the peptide bond involves the participation of a carbon atom of a carboxyl acid group or involves another carbon atom.
  • fuse refers to the act of creating a fused molecule as described above, such as, e.g., a fusion protein generated from the recombinant fusion of genetic regions which when translated produces a single proteinaceous molecule.
  • CD38-binding fusion proteins that may be used in the compositions described herein are described in the art, e.g., in US Patent No. 10544199B2, incorporated herein by reference.
  • the amino acid sequences of an example of an anti-CD38 antibody are provided in Table 1 herein.
  • a CD38-binding fusion protein used in a method described herein comprises an anti- CD38 binder, such as an anti-CD38 antibody or binding domain or fragment thereof.
  • antibody includes, for example, an intact immunoglobulin or an antigen binding portion of an immunoglobulin or an antigen binding protein related or derived from an immunoglobulin. Intact antibody structural units often comprise a tetrameric protein. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one “light” chain (typically having a molecular weight of about 25 kDa) and one “heavy” chain (typically having a molecular weight of about 50- to 70 kDa).
  • Human immunoglobulin light chains may be classified as having kappa or lambda light chains.
  • the antibodies described herein comprise antigen binding domains (e.g., antibody heavy and/or light chains) that generally are based on the IgG class, which has several subclasses, including, but not limited to IgGl, IgG2, IgG3, and IgG4.
  • IgGl has different allotypes with polymorphisms at 356 (D or E), IgG2 and 358 (L or M).
  • the sequences depicted herein use the 356D/358M allotype; however any allotype is included herein and can be used in accordance with the present disclosure.
  • any sequence inclusive of an IgGl Fc domain included herein can have 356E/358L replacing the 356D/358M allotype.
  • the anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a heavy chain comprising a heavy chain variable domain (VH) and a light chain comprising a light chain variable domain (VL).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Each of the loops is referred to as a complementarity-determining region (hereinafter referred to as a “CDR”). Additionally, the variable domains also contain relatively invariant stretches called framework regions (FRs) of 15-30 amino acids separated by CDRs. Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1-CDR1- FR2-CDR2-FR3-CDR3-FR4.
  • an “antibody molecule” refers to two- chain and multi-chain immunoglobulin proteins and glycoproteins.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein is an antibody fragment or antigen binding fragment of an antibody, including, for example, Fab, Fab', F(ab')2, and Fv fragments.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a VH comprising a CDRH1 comprising the amino acid sequence of SEQ ID NO: 1, a CDRH2 comprising the amino acid sequence of SEQ ID NO: 2, and a CDRH3 comprising the amino acid sequence of SEQ ID NO: 3; and a VE comprising a CDRE1 comprising the amino acid sequence of SEQ ID NO: 4, a CDRL2 comprising the amino acid sequence of SEQ ID NO: 5, and a CDRL3 comprising the amino acid sequence of SEQ ID NO: 6.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a set of 6 CDRs that collectively contain up to 10 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid modifications, relative to the 6 CDRs of the anti-CD38 antibody provided in Table 1.
  • the CDRs can be modified in any fashion, as long as the total number of changes in the set of 6 CDRs does not exceed 10 amino acid modifications, with any combination of CDRs being changed; e.g., there may be one change in CDRL1, two in CDRH2, none in CDRH3, etc.
  • each CDR has no more than a single amino acid substitution relative to the corresponding CDR of the anti-CD38 antibody provided in Table 1.
  • amino acid modifications in the CDRH3 are avoided.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a VH comprising the amino acid sequence of SEQ ID NO: 7 and a VL comprising the amino acid sequence of SEQ ID NO: 8.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a VH comprising an amino acid sequence that is at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid sequence of SEQ ID NO: 7 and a VL comprising an amino acid sequence that is at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid sequence of SEQ ID NO: 8.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein is a full-length IgG antibody.
  • each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the immunoglobulin molecules are IgG class IgG4, or a subclass thereof.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises an IgG4 constant region (e.g., a human IgG4 constant region comprising the amino acid sequence of SEQ ID NO: 14).
  • IgG4 constant region refers to a wild-type IgG4 constant region (e.g., a wild-type human IgG4 constant region) or an IgG4 constant region variant (e.g., a human IgG4 constant region variant) or fragment thereof.
  • IgG4 constant region variants (e.g., human IgG4 constant region variants) that may be used in the anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein may, in some embodiments, comprise one or more mutations, e.g., mutations that stabilize the hinge region and/or reduce the toxicity of the antibody.
  • a mutation at position 228 of the IgG4 according to the EU numbering system stabilizes the hinge of IgG4.
  • a mutation at position 228 of the IgG4 constant region according to the EU numbering system results in a proline at position 228.
  • modifications decrease antibody dependent cell cytotoxicity (ADCC).
  • ADCC antibody dependent cell-mediated cytotoxicity
  • FcyRs Fc gamma receptors
  • modifications include, without limitation, mutations at one or more of positions 252, 254, and 256 of the IgG4 constant region according to the EU numbering system.
  • a mutation at position 252 of the IgG4 constant region according to the EU numbering system results in a tyrosine at position 252.
  • a mutation at position 254 of the IgG4 constant region according to the EU numbering system results in a threonine at position 254.
  • a mutation at position 256 of the IgG4 constant region according to the EU numbering system results in a glutamic acid at position 256.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises an IgG4 constant region comprising a mutation at position 228 of the IgG4 constant region according to the EU numbering system.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises an IgG4 constant region comprising the amino acid sequence of SEQ ID NO: 15.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a heavy chain comprising a VH and a human IgG4 constant region, wherein the VH comprises the amino acid sequence of SEQ ID NO: 7 and the IgG4 constant region comprises the amino acid sequence of SEQ ID NO: 15.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO:
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a heavy chain comprising an amino acid sequence at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid sequence of SEQ ID NO: 9.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a light chain comprising a VL and a kappa light constant region, wherein the VL comprises the amino acid sequence of SEQ ID NO: 8.
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a light chain comprising the amino acid sequence of SEQ ID NO:
  • an anti-CD38 antibody of the CD38-binding fusion protein used in a method described herein comprises a light chain comprising an amino acid sequence at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid sequence of SEQ ID NO: 10.
  • a CD38-binding fusion protein used in a method described herein further comprises an anti-CD38 antibody (e.g., the anti-CD38 antibody provided in Table 1) fused to one or more (e.g., one, two) attenuated interferon alpha- 2b protein (e.g., the attenuated interferon alpha- 2b protein is fused to the heavy chain of the anti-CD38 antibody).
  • an anti-CD38 antibody e.g., the anti-CD38 antibody provided in Table 1
  • one or more (e.g., one, two) attenuated interferon alpha- 2b protein e.g., the attenuated interferon alpha- 2b protein is fused to the heavy chain of the anti-CD38 antibody.
  • an attenuated interferon alpha- 2b protein comprises mutations that reduce its potency (e.g., A145D) and/or eliminate O-linked glycosylation of the interferon alpha- 2b protein (e.g., T106A) (in both instances, numbering is relative to the wild type sequence of human interferon-alpha-2b).
  • the potency (e.g., anti-proliferative activity) of attenuated interferon alpha-2b fused to a CD38 binding protein can be determined relative to a non-attenuated IFN- alpha2b (e.g., a wildtype IFN-alpha2b) using an on target (Daudi) assay, as described in USP 11,319,356.
  • a non-attenuated IFN- alpha2b e.g., a wildtype IFN-alpha2b
  • an on target (Daudi) assay as described in USP 11,319,356.
  • An attenuated interferon molecule can be fused to antibodies that specifically bind to CD38 (e.g., an anti-CD38 antibody), as described herein, such that the anti-CD38 antibody may serve as a delivery vehicle for the attenuated interferon to CD38-positive cells with a resulting diminution of off target interferon activity caused by the attenuated interferon molecule.
  • an anti-CD38 antibody e.g., an anti-CD38 antibody
  • the attenuated interferon alpha-2b protein is fused to the heavy chain of the anti-CD38 antibody. In some embodiments, the attenuated interferon alpha-2b protein is fused to the C-terminus of the heavy chain of the anti-CD38 antibody.
  • the CD38-binding fusion protein used in a method described herein comprises a heavy chain and a light chain, wherein the heavy chain comprises the heavy chain of an anti-CD38 antibody fused to an attenuated interferon alpha- 2b protein and wherein the light chain is the light chain of the anti-CD38 antibody.
  • the CD38- binding fusion protein used in a method described herein comprises two heavy chains and two light chains, wherein each heavy chain comprises the heavy chain of an anti-CD38 antibody fused to an attenuated interferon alpha- 2b protein and wherein each light chain is the light chain of the anti-CD38 antibody.
  • the attenuated interferon alpha-2b comprises T106A and A145D mutations relative to a wild type human interferon alpha-2b (e.g., a human interferon alpha- 2b comprising the amino acid sequence of SEQ ID NO: 11).
  • the attenuated interferon alpha- 2b comprises the amino acid of SEQ ID NO: 12.
  • the attenuated interferon alpha- 2b comprises an amino acid sequence at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid of SEQ ID NO: 12.
  • a CD38-binding fusion protein used in a method described herein comprises a heavy chain comprising an amino acid sequence at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid of SEQ ID NO: 13 and a light chain comprising an amino acid sequence at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%) identical to the amino acid of SEQ ID NO: 10.
  • a CD38-binding fusion protein used in a method described herein comprises the amino acid of SEQ ID NO: 13 and a light chain comprising the amino acid of SEQ ID NO: 10.
  • a CD38-binding fusion protein used in a method described herein comprises two heavy chains and two light chains, wherein each heavy comprises the amino acid sequence of SEQ ID NO: 13 and each light chain comprises the amino acid sequence of SEQ ID NO: 10.
  • “Pomalidomide” refers to a thalidomide analog with immunomodulatory, antiangiogenic, and antineoplastic properties. Cellular activities of pomalidomide are mediated through its target cereblon, a component of a cullin ring E3 ubiquitin ligase enzyme complex. Pomalidomide inhibits the proliferation of lenalidomide-resistant multiple myeloma cell lines. Pomalidomide has been shown to enhance T cell- and NK cell-mediated immunity and inhibit production of pro-inflammatory cytokines by monocytes (POMALYST® (pomalidomide) Capsules).
  • POMALYST® pomalidomide
  • Pomalidomide is approved in combination with dexamethasone for patients with multiple myeloma who have received at least 2 prior therapies, including lenalidomide and a proteasome inhibitor, and had demonstrated disease progression on or within 60 days of completion of the last therapy.
  • Lenalidomide refers to a thalidomide analog which inhibits tumor angiogenesis, tumor- secreted cytokines, and tumor proliferation through inhibition of proteasome activity and induction of apoptosis.
  • Lenalidomide (REVLAMID®) has been shown to have direct antitumor effect, activity in inhibition of angiogenesis, and immunomodulation activity.
  • Lenalidomide induces tumor cell apoptosis directly and indirectly by inhibition of bone marrow stromal cell support, by anti- angiogenic and anti-osteoclastogenic effects, and by immunomodulatory activity. Lenalidomide has a broad range of activities that can be exploited to treat many hematologic and solid cancers.
  • proteasome inhibitor refers to a compound that blocks the action of proteasomes, cellular complexes that break down proteins. Proteasome inhibitors have been used in the treatment of cancer. Examples of approved proteasome inhibitors for use in treating multiple myeloma include bortezomib and carfilzomib, which are used as the standard of care treatment for multiple myeloma.
  • Bortezomib has been described, e.g., in US Patent Nos. 6,713,446 and 6,958,319.
  • Bortezomib (VELCADE®) is a proteasome inhibitor that is cytotoxic to a variety of cancer cell types in vitro. Bortezomib causes a delay in tumor growth in vivo in nonclinical tumor models, including multiple myeloma. Bortezomib is indicated for treatment of adult patients with multiple myeloma and in combination with other antimyeloma agents.
  • Carfilzomib has been described, e.g., in US Patent No. 7,232,818.
  • Carfilzomib (KYPROLIS®) covalently irreversibly binds to and inhibits the chymotrypsin-like activity of the 20S proteasome.
  • Carfilzomib displays minimal interactions with non-proteasomal targets, thereby improving safety profiles over bortezomib.
  • Inhibition of proteasome-mediated proteolysis results in a build-up of polyubiquitinated proteins, which may cause cell cycle arrest, apoptosis, and inhibition of tumor growth.
  • the CD38-binding fusion protein is administered once every 4 weeks during a period of administration.
  • agents e.g., the CD38-binding fusion protein described herein, the immunomodulatory drug (e.g., an IMiD such as lenalidomide or pomalidomide), and/or the proteasome inhibitor (e.g., bortezomib or carfilzomib)
  • treatment cycles e.g., one or more treatment cycles
  • each treatment cycle is 4 weeks (i.e., 28 days).
  • a period of administration is up to 5 years (e.g., up to 5 years, up to 4.5 years, up to 4 years, up to 3.5 years, up to 3 years, up to 2.5 years, up to 2 years, up to 23 months, up to 22 months, up to 21 months, up to 20 months, up to 19 months, up to 18 months, up to 17 months, up to 16 months, up to 15 months, up to 14 months, up to 13 months, up to 12 months, up to 11 months, up to 10 months, up to 9 months, up to 8 months, up to 7 months, up to 6 months, up to 5 months, up to 4 months, up to 3 months, up to 2 months, or up to 1 month).
  • a period of administration is more than 2 years (e.g., 2-5, 3-5, 4-5, 2-4, 3-4, or 2-3 years). In some embodiments, a period of administration is until the subject exhibits disease progression. In some embodiments, a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject. In some embodiments, a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • MRD minimal residual disease
  • the CD38-binding fusion protein is administered at 0.5-4 mg/kg (e.g., 0.5-4, 0.5-3.5, 0.5-3, 0.5-2.5, 0.5-2, 0.5-1.5, 0.5-1, 1-4, 1- 3.5, 1-3, 1-2.5, 1-2, 1-1.5, 1.5-4, 1.4-3.5, 1.5-3, 1.5-2.5, 1.5-2, 2-4, 2-3.5, 2-3, 2-2.5, 2.5-4, 2.5- 3.5, 2.5-3, 3-4, 3-3.5, or 3.5-4 mg/kg) of the subject.
  • 0.5-4 mg/kg e.g., 0.5-4, 0.5-3.5, 0.5-3, 0.5-2.5, 0.5-2, 0.5-1.5, 0.5-1, 1-4, 1- 3.5, 1-3, 1-2.5, 1-2, 1-1.5, 1.5-4, 1.4-3.5, 1.5-3, 1.5-2.5, 1.5-2, 2-4, 2-3.5, 2-3, 2-2.5, 2.5-4, 2.5- 3.5, 2.5-3, 3-4, 3-3.5, or 3.5-4 mg/kg
  • the CD38-binding fusion protein in a method described herein, is administered at 0.75-3 mg/kg (e.g., 0.75- 3, 1-3, 1.2-2.8, 1.4-2.6, 1.6-2.4, 1.8-2.2, or 1.9-2.1 mg/kg) of the subject. In some embodiments, in a method described herein, the CD38-binding fusion protein is administered at about 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 3.5, or 4 mg/kg of the subject. In some embodiments, in a method described herein, the CD38-binding fusion protein is administered at about 1 mg/kg of the subject.
  • the CD38-binding fusion protein is administered at a dose of 50-250 mg (e.g., 50-250, 50-200, 50-150, 50-100, 60-250, 60-200, 60-150, 60-100, 100-250, 100-200, 100-150, 150-250, 150-200, or 200-250 mg) to the subject regardless of the subject’s weight.
  • the CD38-binding fusion protein is administered at a dose of 60-240 mg (e.g., 60-240, 80-240, 100-200, or 120-160 mg) to the subject regardless of the subject’s weight.
  • the CD38-binding fusion protein in a method described herein, is administered at a dose of about 60 mg, 80 mg, 120 mg, or 240 mg to the subject regardless of the subject’s weight. In some embodiments, in a method described herein, the CD38-binding fusion protein is administered at a dose of about 80 mg to the subject regardless of the subject’s weight.
  • the CD38-binding fusion protein is administered at 0.75-3 mg/kg (e.g., about 0.75, 1, 1.5, 2, 2.5, or 3 mg/kg) of the subject or at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg regardless of the subject’s weight) according to treatment cycles (e.g., 4-week (i.e., 28-day) treatment cycles) during a period of administration (e.g., any one of the period of administration described herein).
  • treatment cycles e.g., 4-week (i.e., 28-day) treatment cycles
  • the CD38-binding fusion protein is administered at 0.75-3 mg/kg (e.g., about 0.75, 1, 1.5, 2, 2.5, or 3 mg/kg) of the subject or at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg regardless of the subject’s weight) once in each 4-week treatment cycle (e.g., on day 1 of each treatment cycle).
  • the CD38-binding fusion protein is administered intravenously (e.g., via intravenous injection or infusion).
  • the immunomodulatory drug administered to the subject is pomalidomide (e.g., according to any known dose and dosing regimen for pomalidomide).
  • the subject in a method described herein, is administered 2-6 mg (e.g., 2-6, 2-5, 2-4, 2-3, 3-6, 3-5, 3-4, 4-6, 4-5, or 5-6 mg) of pomalidomide daily.
  • the subject in a method described herein, the subject is administered 2, 3, 4, 5, or 6 mg of pomalidomide daily.
  • the subject is administered 4 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle during a period of administration.
  • pomalidomide is administered orally (e.g., as a capsule).
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, and the immunomodulatory drug pomalidomide, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, pomalidomide is administered (e.g., orally as a capsule) at a dose of 2-4 mg daily for the first 21 days of each 4-week treatment cycle during a period of administration.
  • the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle
  • pomalidomide is administered (e.g., orally as a
  • the method comprises administering 2-3 mg of pomalidomide. In some embodiments, the method comprises administering 3-4 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, the method comprises administering 2 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, the method comprises administering 3 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, the method comprises administering 4 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject.
  • a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • the immunomodulatory drug administered to the subject is lenalidomide (e.g., according to any known dose and dosing regimen for lenalidomide). In some embodiments, in a method described herein, the subject is administered 5-15 mg (e.g., 5-15, 5-12, 5-9, 5-6, 8-15, 8-12, 8-9, 9-15, 9-12, or 12-15 mg) of lenalidomide daily.
  • the subject in a method described herein, is administered about 5, 6, 7, 8, 9, 10, 11, 12, 13 ,14, or 15 mg of lenalidomide daily.
  • the method comprises administering lenalidomide at a dose of 5-15 mg (e.g., 10 mg) up to daily for a 4-week treatment cycle.
  • the method comprises administering lenalidomide at a dose of 5-15 mg (e.g., 10 mg) daily for a 4-week treatment cycle.
  • the subject in a method described herein, is administered 10 mg of lenalidomide daily for 28 days of each 4-week treatment cycle during a period of administration (i.e., lenalidomide is administered every day of each 4-week treatment cycle).
  • lenalidomide is administered orally (e.g., as a capsule).
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, and the immunomodulatory drug lenalidomide, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, lenalidomide is administered (e.g., orally as a capsule) at a dose of 10 mg daily for the first 28 days of each 4-week treatment cycle during a period of administration.
  • the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle
  • lenalidomide is administered (e.g., orally as a capsule
  • the dose of lenalidomide is increased to 15 mg daily if tolerated.
  • a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject.
  • a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative).
  • a period of administration lasts the remainder of the subject’s lifetime.
  • the proteasome inhibitor administered to the subject is bortezomib.
  • bortezomib is administered according to any known dose and dosing regimen for bortezomib.
  • bortezomib is administered at a dose based on the subject’s body surface area.
  • bortezomib is administered at a dose of 1-1.5 mg/m 2 (e.g., 1-1.5, 1-1.4, 1-1.3, 1-1.2, 1-1.1, 1.1-1.5, 1.1-1.4, 1.1-1.3, 1.1- 1.2, 1.2-1.5, 1.2-1.4, 1.2-1.3, 1.3-1.5, 1.3-1.4, or 1.4-1.5 mg/m 2 ). In some embodiments, in a method described herein, bortezomib is administered at a dose of about 1, 1.1, 1.2, 1.3, 1.4, or 1.5 mg/m 2 .
  • the method comprises administering bortezomib at a dose of 1-2 mg/m 2 weekly for a 4-week treatment cycle for 1, 2, 3, 4, 5, 6, 7, 8, or 10 treatments cycles and then administering bortezomib at a dose of 1-2 mg/m 2 every two weeks for any remaining treatment cycles.
  • bortezomib is administered at a dose of about 1.3 mg/m 2 on days 1, 8, 15 for eight 4-week treatment cycles and on days 1 and 8 for remaining 4-week treatment cycles during a period of administration.
  • bortezomib is administered subcutaneously (e.g., via subcutaneous injection).
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, and the proteasome inhibitor bortezomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, and bortezomib is administered (e.g., via subcutaneous injection) at a dose of about 1-2 mg/m 2 on days 8, 15 and 22 for eight 4-week treatment cycles and on days 8 and 22 for remaining 4-week treatment cycles during a period of administration.
  • the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment
  • a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject. In some embodiments, a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, and the proteasome inhibitor bortezomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, and bortezomib is administered (e.g., via subcutaneous injection) at a dose of about 1.3 mg/m 2 on days 8, 15 and 22 for eight 4-week treatment cycles and on days 8 and 22 for remaining 4-week treatment cycles during a period of administration.
  • the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment
  • a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject. In some embodiments, a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • the proteasome inhibitor administered to the subject is carfilzomib.
  • carfilzomib is administered according to any known dose and dosing regimen for carfilzomib.
  • carfilzomib is administered at a dose based on the subject’s body surface area.
  • bortezomib is administered at a dose of 10-30 mg/m 2 (e.g., 10-30, 10-25, 10-20, 10-15, 15-30, 15-25, 15-20, 20-30, 20-25, or 25-30 mg/m 2 ).
  • carfilzomib in a method described herein, is administered at a dose of about 10, 15, 20, 25, 30 mg/m 2 . In some embodiments, in a method described herein, carfilzomib is administered at a dose of up to 70 mg/m 2 (e.g., up to 70, up to 60, or up to 50 mg/m 2 ). In some embodiments, in a method described herein, carfilzomib is administered at a dose of about 70 mg/m 2 .
  • carfilzomib in a method described herein, is administered at a dose of about 20 mg/m 2 on day 1 of cycle 1 (i.e., the first 4-week treatment cycle during a period of administration), and at a dose of up to 70 mg/m 2 on days 8 and 15 for remaining 4-week treatment cycles during a period of administration.
  • carfilzomib in a method described herein, is administered intravenously (e.g., intravenous injection or infusion).
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, and the proteasome inhibitor carfilzomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, carfilzomib is administered (e.g., via intravenous infusion) at a dose of about 20 mg/m 2 on day 1 and at a dose of up to 70 mg/m 2 on days 8 and 15 of cycle 1 (i.e., the first 4- week treatment cycle during a period of administration), and at a dose of up to 70 mg/m 2 for remaining 4-week treatment cycles during a period of administration.
  • the CD38-binding fusion protein is administered (e.g., via intravenous infusion
  • a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject. In some embodiments, a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • a method of treating a CD38-expressing cancer described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject in need thereof a CD38-binding fusion protein described herein, pomalidomide, and bortezomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, pomalidomide is administered (e.g., orally as a capsule) at a dose of 4 mg daily for up to the first 21 days of each 4-week treatment cycle (e.g., depending on adverse patient side effects), and bortezomib is administered (e.g., via subcutaneous injection) at a dose of 1-2 mg/m 2 (e.g., 1.3 mg/m 2 ) on days 1, 8, 15 for eight 4-week treatment cycles and on days 1
  • a method described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject having multiple myeloma the CD38-binding fusion protein described herein, pomalidomide, and bortezomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, pomalidomide is administered (e.g., orally as a capsule) at a dose of 2-4 mg daily for the first 21 days of each 4-week treatment cycle, and bortezomib is administered (e.g., via subcutaneous injection) at a dose of about 1.3 mg/m 2 on days 8, 15 and 22 for eight 4-week treatment cycles and on days 8 and 22 for remaining 4-week treatment cycles during a period of administration.
  • the CD38-binding fusion protein is administered (e.g.,
  • the method comprises administering 2-3 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments daily for the first 21 days of each 4-week treatment cycle, the method comprises administering 2 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, the method comprises administering 3 mg of pomalidomide daily for the first 21 days of each 4-week treatment cycle. In some embodiments, a period of administration is until the subject exhibits disease progression. In some embodiments, a period of administration lasts until an event meeting the criteria for discontinuing the treatment is met (e.g., unacceptable toxicity is observed) for the subject. In some embodiments, a period of administration ends when no cancer cells are detected in the subject (e.g., when the subject is minimal residual disease (MRD)-negative). In some embodiments, a period of administration lasts the remainder of the subject’s lifetime.
  • MRD minimal residual disease
  • the method comprises administering an CD38-binding fusion protein comprising: (i) a first anti-CD38 antibody comprising a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 8; and (ii) an attenuated interferon alpha- 2b comprising an amino acid sequence of SEQ ID NO: 12.
  • a method of treating a CD38-expressing cancer described herein comprises administering, according to a 4-week treatment cycle during a period of administration, to a subject in need thereof a CD38-binding fusion protein described herein, pomalidomide, and carfilzomib, wherein the CD38-binding fusion protein is administered (e.g., via intravenous infusion) at a dose of 60-240 mg (e.g., about 60, 80, 120, or 240 mg) on day 1 of each 4-week treatment cycle, pomalidomide is administered (e.g., orally as a capsule) at a dose of 4 mg daily for the first 21 days of each 4-week treatment cycle, and carfilzomib is administered at a dose of about 20 mg/m 2 on day 1 of cycle 1 (i.e., the first 4-week treatment cycle during a period of administration), and at a dose of up to 70 mg/m 2 on days 8 and 15 for remaining 4-week treatment cycles during a period of administration.
  • the subject in any one of the methods described herein, is receiving or has received treatment with a steroid, e.g., dexamethasone.
  • a steroid e.g., dexamethasone.
  • the subject in any one of the methods described herein, is not receiving or has not received treatment with a steroid, e.g., dexamethasone.
  • a steroid e.g., dexamethasone.
  • the CD38-binding fusion protein is formulated in a composition for administration.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a buffer (e.g., a histidine/histidine-HCl buffer), a tonicity agent (e.g., arginine- HCL), a stabilizer (e.g., sucrose), and a surfactant (e.g., polysorbate such as polysorbate 80).
  • a buffer e.g., a histidine/histidine-HCl buffer
  • a tonicity agent e.g., arginine- HCL
  • a stabilizer e.g., sucrose
  • a surfactant e.g., polysorbate such as polysorbate 80.
  • a composition described herein has a pH between 6.1-7.1 (e.g., 6.6) and comprises a CD38-binding fusion protein at a concentration of 8-12 mg/mL (e.g., 10 mg/ml), histidine/histidine-HCl at a concentration of 40-60 mM (e.g., 50 mM), arginine-HCL at a concentration of 75-125 mM (e.g., 100 mM), sucrose at a concentration of 30-80 mg/ml (e.g., 50 mg/ml), and polysorbate 80 at a 0.1-0.3 mg/ml (e.g., 0.2 mg/ml).
  • a CD38-binding fusion protein at a concentration of 8-12 mg/mL (e.g., 10 mg/ml)
  • histidine/histidine-HCl at a concentration of 40-60 mM (e.g., 50 mM)
  • arginine-HCL at a concentration
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a CD38-binding fusion protein at a concentration that does not exceed 100 mg/ml. In some embodiments, a composition comprising the CD38- binding fusion protein used in a method described herein comprises a CD38-binding fusion protein at a concentration of 8-12 mg/ml.
  • a composition comprising the CD38- binding fusion protein used in a method described herein may comprise a CD38-binding fusion protein at a concentration of 8-12 mg/ml, 8-11.5 mg/ml, 8-11 mg/ml, 8-10.5 mg/ml, 8-10 mg/ml, 8-9.5 mg/ml, 8-9 mg/ml, 8-8.5 mg/ml, 8.5-12 mg/ml, 8.5-11.5 mg/ml, 8.5-11 mg/ml, 8.5-10.5 mg/ml, 8.5-10 mg/ml, 8.5-9.5 mg/ml, 8.5-9 mg/ml, 9-12 mg/ml, 9-11.5 mg/ml, 9-11 mg/ml, 9-10.5 mg/ml, 9-10 mg/ml, 9-9.5 mg/ml, 9.5-12 mg/ml, 9.5-11.5 mg/ml, 9.5-11 mg/ml, 9.5-10.5 mg/ml, 9.5-10 mg/ml, 10-12 mg/ml, 10-12 mg
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a CD38-binding fusion protein at a concentration of about 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, 10, 10.1, 10.2, 10.3, 10.4, 10.5, 10.6, 10.7, 10.8, 10.9, 11, 11.1, 11.2, 11.3, 11.4, 11.5, 11.6, 11.7, 11.8, 11.9, or 12 mg/ml.
  • a composition comprising the CD38- binding fusion protein used in a method described herein comprises a CD38-binding fusion protein at a concentration of about 10 mg/ml.
  • a composition comprising the CD38-binding fusion protein used in a method described herein has a pH of 5.5-7.5.
  • a composition comprising the CD38-binding fusion protein used in a method described herein may have a pH of 5.5-7.5, 5.5- 7, 5.5-6.5, 5.5-6, 6-7.5, 6-7, 6-6.5, 6.5-7.5, 6.5-7, or 7-7.5.
  • a composition described herein has a pH of about 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, or 7.5.
  • a composition comprising the CD38-binding fusion protein used in a method described herein has a pH of about 6.1-7.1 (e.g., 6.1-7.1, 6.2-7, 6.3-6.9, 6.4-6.8, or 6.5-6.7). In some embodiments, a composition comprising the CD38-binding fusion protein used in a method described herein has a pH of about 6.6.
  • a composition comprising the CD38-binding fusion protein used in a method as described herein further comprises a buffer (e.g., a histidine/histidine-HCl buffer), a tonicity agent (e.g., arginine-HCL), a stabilizer (e.g., sucrose), and a surfactant (e.g., polysorbate such as polysorbate 80).
  • a buffer e.g., a histidine/histidine-HCl buffer
  • a tonicity agent e.g., arginine-HCL
  • a stabilizer e.g., sucrose
  • a surfactant e.g., polysorbate such as polysorbate 80.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a buffer comprising histidine and histidine-HCL.
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of 10-120 mM (e.g., 10-120 mM, 20-110 mM, 30-100 mM, 40-90 mM, 50-80 mM, or 60-70 mM).
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of 12.5-107.5 mM.
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of 15-50 mM (e.g., about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, or about 50 mM).
  • 15-50 mM e.g., about 15 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, or about 50 mM.
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57,
  • the relative amount of histidine and histidine-HCL may be adjusted, e.g., to achieve a desired pH, while maintaining the histidine concentration in the composition, as described herein.
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of about 15 mM (e.g., when the composition comprises a buffer comprising histidine at a concentration of 7.5 mM and histidine-HCL at a concentration of 7.5 mM).
  • the histidine and histidine-HCL balance results in a final histidine concentration in the composition of about 50 mM (e.g., when the composition comprises a buffer comprising histidine at a concentration of 40 mM and histidine-HCL at a concentration of 10 mM).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a tonicity agent comprising arginine-HCL.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises arginine-HCL at a concentration of 50-125 mM (e.g., 50-125 mM, 60-120 mM, 70-110 mM, or 80-100 mM, 75-125 mM, 95-105 mM, or 97.5-102.5 mM).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises arginine-HCL at a concentration of about 50 mM, 55 mM, 60 mM, 65 mM, 70 mM, 75 mM, 80 mM, 85 mM, 90 mM, 95 mM, 100 mM, 105 mM, 110 mM, 115 mM, or 120 mM.
  • a composition comprising the CD38- binding fusion protein used in a method described herein comprises arginine-HCL at a concentration of about 100 mM.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a stabilizer.
  • the stabilizer is a sugar.
  • the stabilizer is sucrose.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of 3-10 % w/v (equivalent to 30-100 mg/ml).
  • a composition described herein may comprise sucrose at a concentration of 3-10 % w/v, 3-9 % w/v, 3-8 % w/v, 3-7 % w/v, 3-6 % w/v, 3-5 % w/v, 3-4 % w/v, 3-10 % w/v, 3-9 % w/v, 3-8 % w/v, 3-7 % w/v, 3-6 % w/v, 3-5 % w/v, 3-4 % w/v, 4-10 % w/v, 4-9 % w/v, 4-8 % w/v, 4-7 % w/v, 4-6 % w/v, 4-5 % w/v, 5-10 % w/v, 5-9 % w/v, 5-8 % w/v, 5-7 % w/v, 5-6 % w/v, 6-10 % w/v, 6-9 w/
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 3% w/v (equivalent to 30 mg/mL), 3.5% w/v (equivalent to 35 mg/mL), 4% w/v (equivalent to 40 mg/mL), 4.5% w/v (equivalent to 45 mg/mL), 5% w/v (equivalent to 50 mg/mL), 5.5% w/v (equivalent to 55 mg/mL), 6% w/v (equivalent to 60 mg/mL), 6.5% w/v (equivalent to 65 mg/mL), 7% w/v (equivalent to 70 mg/mL), 7.5% w/v (equivalent to 75 mg/mL), 8% w/v (equivalent to 80 mg/mL), 8.5% w/v (equivalent to 85 mg/mL), 9w/v (equivalent to 90 mg/mL),
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 4%-8% w/v (equivalent to 40-80 mg/mL). In some embodiments, a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 4%-7% w/v (equivalent to 40-70 mg/mL). In some embodiments, a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 4%- 6% w/v (equivalent to 40-60 mg/mL).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 4.5%-5.5% w/v (equivalent to 45-55 mg/mL). In some embodiments, a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 4% w/v, 5% w/v, 6% w/v, 7% w/v, or 8 % w/v (equivalent to 40 mg/ml, 50 mg/ml, 60 mg/ml, 70 mg/ml or 80 mg/ml, respectively). In some embodiments, a composition comprising the CD38-binding fusion protein used in a method described herein comprises sucrose at a concentration of about 5% w/v (equivalent to 50 mg/ml).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a surfactant.
  • the surfactant is a polysorbate.
  • the surfactant is a polysorbate 80 (PS 80).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises PS80 at a concentration of 0.005-0.03 % w/v (equivalent to 0.05- 0.3 mg/ml).
  • a composition comprising the CD38-binding fusion protein used in a method described herein may comprise PS80 at a concentration of 0.005-0.03 % w/v, 0.005- 0.025 % w/v, 0.005-0.02 % w/v, 0.005-0.015 % w/v, 0.005-0.01% w/v, 0.01-0.03 % w/v, 0.01- 0.025 % w/v, 0.01-0.02 % w/v, 0.01-0.015 % w/v, 0.015-0.03 % w/v, 0.015-0.025 % w/v, 0.015-0.02 % w/v, 0.02-0.03 % w/v, 0.02-0.025 % w/v, 0.02-0.03 % w/v, 0.02-0.025 % w/v, 0.02-0.03 % w/v, 0.02-0.025 % w/v, 0.0
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises PS 80 at a concentration of about 0.007% w/v (equivalent to 0.07 mg/mL), 0.008% w/v (equivalent to 0.08 mg/mL), 0.009% w/v (equivalent to 0.09 mg/mL), 0.01% w/v (equivalent to 0.1 mg/mL), 0.011% w/v (equivalent to 0.11 mg/mL), 0.012% w/v (equivalent to 0.12 mg/mL), 0.013% w/v (equivalent to 0.13 mg/mL), 0.014% w/v (equivalent to 0.14 mg/mL), 0.015% w/v (equivalent to 0.15 mg/mL), 0.016% w/v (equivalent to 0.16 mg/mL), 0.017% w/v (equivalent to 0.17 mg/mL), 0.018% w/v (equi
  • a composition described herein comprises PS80 at a concentration of about 0.01%-0.03% w/v (equivalent to 0.1-0.3 mg/mL).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises PS80 at a concentration of about 0.015%-0.025% w/v (equivalent to 0.15-0.25 mg/mL).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises PS 80 at a concentration of about 0.02% w/v (equivalent to 0.2 mg/ml).
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a CD38-binding fusion protein (e.g., a CD38-binding fusion protein as provided in Table 1) at a concentration of 8.5-11.5 mg/ml (e.g., 10 mg/ml), histidine (e.g., composed of histidine and histidine-HCL) at a concentration of 15-60 mM (e.g., 15 mM, 20 mM, 30 mM, 40 mM, or 50 mM), arginine-HCL at a concentration of 80-120 mM (e.g., 100 mM), sucrose at a concentration of 3-8% w/v (e.g., 5% w/v), and PS80 at a concentration of 0.01-0.03% w/v (e.g., 0.02% w/v), and wherein the composition is at a pH of 5.5-7.5 (e.g.,
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a CD38-binding fusion protein (e.g., a CD38-binding fusion protein as provided in Table 1) at a concentration of 10 mg/ml, histidine (e.g., composed of histidine and histidine-HCL) at a concentration of 50 mM, arginine-HCL at a concentration of 100 mM, sucrose at a concentration of 5% w/v, and PS80 at a concentration of 0.02% w/v, and wherein the composition is at a pH of 6.6.
  • the CD38-binding fusion protein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 13 and a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • a composition comprising the CD38-binding fusion protein used in a method described herein comprises a CD38-binding fusion protein (e.g., a CD38-binding fusion protein as provided in Table 1) at a concentration of 10 mg/ml, histidine (e.g., composed of histidine and histidine-HCL) at a concentration of 15 mM, arginine-HCL at a concentration of 100 mM, sucrose at a concentration of 5% w/v, and PS80 at a concentration of 0.02% w/v, and wherein the composition is at a pH of 6.
  • the CD38-binding fusion protein comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 13 and a light chain comprising the amino acid sequence of SEQ ID NO: 10.
  • a composition comprising the CD38-binding fusion protein used in a method described herein is an aqueous solution.
  • a composition described herein (e.g., in a form of aqueous solution or in lyophilized form) is stored in dosage unit form.
  • a lyophilized form of a composition described herein is stored for at least 2 months, at least 4 months, at least 6 months, at least 1 year, at least 2 years, or at least 3 years.
  • a composition described herein (e.g., in a form of aqueous solution or in lyophilized form) is stored frozen.
  • a method described herein is effective in treating a cancer in a patient. Treating may include, for example, inhibiting or reducing proliferation of CD38- positive cells in the cancer and/or inducing apoptosis of CD38-positive cells in the cancer.
  • the terms “subject” and “patient” are used interchangeably and include any mammals, including companion and farm mammals, as well as rodents, including mice, rabbits, and rats, and other rodents. Non-human primates, such as Cynomolgus monkeys, are more preferred, and human beings are highly preferred.
  • the subject is a human.
  • the subject is a human adult (e.g., more than 18 years old, including 18 years old).
  • the subject is a non-adult human (e.g., less than 18 years old).
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof or reducing the likelihood of a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development or progression; and (c) relieving the disease, i.e., causing regression of the disease and/or relieving one or more disease symptoms. “Treatment” is also meant to encompass delivery of an agent in order to provide for a pharmacologic effect, even in the absence of a disease or condition.
  • the cancer is a CD38-expressing cancer. In some embodiments, the cancer is not a CD38-expressing cancer. In some embodiments, the cancer is multiple myeloma (MM). In some embodiments, the subject has not received any prior lines of MM therapy. In some embodiments, the subject has not received any prior anti-CD38-based therapies (e.g., using daratumumab).
  • the subject has received prior lines of therapy (e.g., one or more prior lines of therapy) for multiple myeloma, including at least one proteasome inhibitor, at least one anti-CD38 monoclonal antibody (mAb) drug, and/or at least one immunomodulatory drug (e.g., an IMiD).
  • the subject is refractory to a proteasome inhibitor, an immunomodulatory drug (e.g., an IMiD), or an anti-CD38 antibody therapy regardless of prior lines of therapy.
  • the subject demonstrated disease progression with the one or more prior lines of therapy.
  • the subject has undergone standard of care (SOC) treatment for MM.
  • subject has undergone autologous stem cell transplant (ASCT) and is responsive to such treatment.
  • SOC standard of care
  • ASCT autologous stem cell transplant
  • the subject suffers from Relapsed or Refractory Multiple Myeloma (RRMM).
  • RRMM Refractory Multiple Myeloma
  • the subject with RRMM has failed treatment with, is intolerant to, or is not a candidate for available therapies that are known to confer clinical benefit in RRMM patients.
  • the RRMM subject has received at least three prior lines of MM therapy and is refractory or intolerant to at least one proteasome inhibitor (PI) based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • the prior lines of MM therapy can include one or more anti-CD38 therapy, including but not limited to daratumumab.
  • the RRMM subject has received at least three prior lines of MM therapy including daratumumab, and is relapsed or refractory to daratumumab, at least one proteasome inhibitor (PI) based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • MM therapy including daratumumab, and is relapsed or refractory to daratumumab, at least one proteasome inhibitor (PI) based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • PI proteasome inhibitor
  • IMiD immunomodulatory drug
  • the RRMM subject has received at least three prior lines of MM therapy, and is refractory to at least one proteasome inhibitor (PI) based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid- based therapy.
  • the prior lines of MM therapy do not include any anti-CD38 therapy.
  • the RRMM subject has received at least two prior lines of MM therapy, wherein one of these two lines includes a combination of a PI based therapy and an immunomodulatory drug (e.g., an IMiD) based therapy, and the subject is refractory to at least one PI based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • One or more of the prior lines of MM therapy can include an anti-CD38 therapy, including but not limited to daratumumab.
  • the RRMM subject has received at least two prior lines of MM therapy, wherein one of these two lines includes a combination of a PI based therapy and an immunomodulatory drug (e.g., an IMiD) based therapy, and the other line includes daratumumab, and the subject is relapsed or refractory to daratumumab, at least one PI based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • an immunomodulatory drug e.g., an IMiD
  • the RRMM subject has received at least two prior lines of MM therapy, wherein one of these two lines includes a combination of a PI based therapy and an immunomodulatory drug (e.g., an IMiD) based therapy, and the subject is refractory to at least one PI based therapy, at least one immunomodulatory drug (e.g., an IMiD) based therapy, and optionally at least one steroid-based therapy.
  • the prior lines of MM therapy do not include any anti-CD38 therapy.
  • the RRMM subject has received prior lines of anti-CD38 therapy, including but not limited to daratumumab, and the subject is relapsed or refractory to the anti-CD38 therapy at any time during treatment with the methods described herein.
  • Example 1 A Phase lb Open-label Study to Evaluate the Safety and Tolerability of Intravenous CD 38 -binding fusion protein as Part of Combination Therapy in Adult Patients With Multiple Myeloma This study aims to determine proper dosing and administration approaches of a CD38- binding fusion protein in combination with other compounds for treating multiple myeloma.
  • CD38 is a multifunctional ectoenzyme involved in cell adhesion and transmembrane signaling. It is overexpressed in various tumor types and is believed to play a role in tumor cell migration and metastasis. CD38 is an approximately 45 kDa transmembrane glycoprotein expressed by immature hematopoietic cells, downregulated in mature cells, and re-expressed at higher levels by activated lymphocytes, such as T cells, B cells, dendritic cells, and natural killer (NK) cells.
  • activated lymphocytes such as T cells, B cells, dendritic cells, and natural killer (NK) cells.
  • the CD38-binding fusion protein evaluated in this study is a recombinant humanized immunoglobulin (Ig) G4 anti-CD38 monoclonal antibody fused to 2 attenuated interferonalpha 2b (IFNa2b) moieties.
  • the amino acid sequences of the CD38-binding fusion protein are provided in Table 1 (heavy chain fused to attenuated interferon alpha-2b, i.e., SEQ ID NO: 13, and light chain, i.e., SEQ ID NO: 10).
  • the CD38-binding fusion protein was produced by recombinant DNA technology in a mammalian cell expression system and was purified by a process that included specific viral inactivation and removal steps.
  • CD38 antibody portion of CD38-binding fusion protein directs the attenuated IFNa2b portion to CD38-expressing (CD38+) cells, thus achieving a high local concentration of IFNa2b at the surface of these target cells.
  • CD38-negative cells the attenuation resulted is approximately 130,000-fold reduced potency compared with IFNa2b.
  • the CD38-binding fusion protein has a high binding affinity (dissociation constant [KD]) for human and cynomolgus CD38, with a KD of 168 pM and 1.25 nM, respectively.
  • CD38-binding fusion protein potently inhibits proliferation of CD38+ multiple myeloma (MM) cells (half-maximal inhibitory concentration [IC50] 19.9 pM), whereas potency on CD38- negative cells is approximately 2500-fold lower.
  • the antibody portion of CD38-binding fusion protein is an IgG4 isotype and therefore has limited effector capacity to induce antibodydependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis, or complement activation against normal CD38+ cells.
  • CD38-binding fusion protein does not modulate the adenosine dipho sphate-ribosyl cyclase activity of CD38.
  • the main aims of this study are to evaluate side effects, safety, and tolerability of a CD38-binding fusion protein in combination therapy and to determine the recommended phase 2 dose (RP2D) of combination therapy with the CD38-binding fusion protein and how often administration occurs.
  • the dose of the CD38-binding fusion protein is increased incrementally over time until the highest dose that does not cause harmful side effects is found. Participants are given the CD38-binding fusion protein through intravenous infusion.
  • the study determines the safety, tolerability, and the recommended dose of CD38- binding fusion protein in combination with lenalidomide in participants with newly diagnosed multiple myeloma (NDMM), and in combination with pomalidomide, or bortezomib, or carfilzomib in participants with relap sed/refractory multiple myeloma (RRMM).
  • NDMM newly diagnosed multiple myeloma
  • pomalidomide or bortezomib
  • carfilzomib in participants with relap sed/refractory multiple myeloma
  • Group 1 corresponds to newly diagnosed multiple myeloma (NDMM) patients in maintenance therapy.
  • Group 2 corresponds to RRMM patients who have received at least 2 prior lines of therapy, including at least 1 proteasome inhibitor (PI), at least 1 CD38 monoclonal antibody (mAb) drug, and at least 1 immunomodulatory drug (IMiD), and/or who were refractory to a PI, an IMiD, and a CD38 therapy regardless of prior lines of therapy; and who demonstrated disease progression with the last therapy.
  • Group 3 corresponds to RRMM patients who have received 1 to 3 prior lines of antimyeloma therapy including at least 1 PI, 1 IMiD, and 1 anti-CD38 mAb and who are not refractory to the combination partners.
  • Participants must have received at least 2 prior lines of therapy, including at least 1 PI, at least 1 CD38 monoclonal antibody (mAb) drug, and at least 1 immunomodulatory drug (IMiD), or who are triple refractory to a PI, and IMiD, and an anti-CD38 mAb drug regardless of the number of prior line(s) of therapy.
  • mAb CD38 monoclonal antibody
  • IiD immunomodulatory drug
  • MM have a diagnosis of primary amyloidosis, Waldenstrom disease, monoclonal gammopathy of undetermined significance or smoldering MM per IMWG criteria or standard diagnostic criteria, plasma cell leukemia POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal protein, and skin changes), or lymphoplasmacytic lymphoma;
  • hepatitis B defined by a positive test for hepatitis B surface antigen [HBsAg]
  • seropositive for hepatitis B (defined by a positive test for hepatitis B surface antigen [HBsAg])
  • participants with resolved infection that is, participants who are HBsAg negative but positive for antibodies to hepatitis B core antigen and/or antibodies to hepatitis B surface antigen [anti-HBs]
  • PCR real-time polymerase chain reaction
  • HBV hepatitis B virus
  • the study groups receive administration regimens as described below in Table 2.
  • CD38-binding fusion protein infusion intravenously (IV), once on Day 1, once every 4 ⁇
  • Primary outcome measures of the study include:
  • DLT dose-limiting toxicities
  • hematologic toxicity nonfebrile grade 4 neutropenia/grade greater than or equal to (>) 3 febrile neutropenia;
  • PFS Progression Free Survival
  • PD Progression Free Survival
  • PD may be defined by an increase of >25 percent (%) from lowest response value in any one or more of the following: serum M-component increase >0.5 gram per deciliter (g/dL) or urine M-component increase >200 milligram (mg)/24-hour; difference between involved and uninvolved free light chains (FLC) levels increase must be greater than (>) 10 milligram per deciliter (mg/dL); bone marrow plasma cell >10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; or development of hypercalcemia that can be attributed solely to plasma cell proliferative disorder.
  • difference between involved and uninvolved free light chains (FLC) levels increase must be greater than (>) 10 milligram per deciliter (mg/dL)
  • ORR Overall response rate of up to approximately 5 years.
  • ORR is defined as the percentage of participants who achieved a partial response rate (PR) or better during the study as defined by IMWG Uniform Response Criteria.
  • PR may be determined by >50% reduction of serum M-protein and >90% reduction in urine M-protein or less than ( ⁇ ) 200 mg/24 hour, or >50% decrease in uninvolved FLC or >50% reduction in plasma cells. At baseline, a >50% decrease in size of soft tissue plasmacytomas is required.
  • DOR Duration of response
  • PR may be determined by >50% reduction of serum M-protein and >90% reduction in urine M-protein or ⁇ 200 mg/24 hour, or >50% decrease in uninvolved FLC or >50% reduction in plasma cells. At baseline, a >50% decrease in size of soft tissue plasmacytomas is required.
  • PD may be determined by an increase of >25% from lowest response value in any one or more of the following: serum M-component increase >0.5 g/dL or urine M-component increase >200 mg/24-hour; difference between involved and uninvolved FLC levels increase must be >10 mg/dL; bone marrow plasma cell >10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; development of hypercalcemia that can be attributed solely to plasma cell proliferative disorder.
  • OS Overall survival rates in Groups 2 and 3 of up to approximately 5 years. OS is defined as the time from the first dose of administration to the date of death, due to any cause. Participants without documentation of death at the time of analysis will be censored at the date last known to be alive.
  • TTP Time to progression
  • PD may be determined by an increase of >25% from lowest response value in any one or more of the following: serum M-component increase >0.5 g/dL or urine M-component increase >200 mg/24-hour; difference between involved and uninvolved FLC levels increase must be >10 mg/dL; bone marrow plasma cell >10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; development of hypercalcemia that can be attributed solely to plasma cell proliferative disorder.
  • TTNT Time to next treatment in Groups 2 and 3 over a 5-year time frame.
  • TTNT is defined as the time from the date of first dose administration to the date of the first dose initiation of the next line of antineoplastic therapy, for any reason.
  • DCR Disease control rate in Groups 2 and 3 over a 5-year time frame.
  • DCR is defined as the percentage of participants who achieved a stable disease (SD) or better during the study as defined by IMWG Uniform Response Criteria. SD is defined as no known evidence of progressive disease or new bone lesions.
  • CBR Clinical benefit rate
  • sCR complete response
  • VGPR very good partial response
  • PR partial response
  • IMWG minimal response per IMWG criteria.
  • CR may be determined by negative immunofixation of serum and urine, disappearance of any soft tissue plasmacytomas, ⁇ 5% plasma cells in bone marrow; in participants for whom only measurable disease is by serum FLC level, normal FLC ratio of 0.26 to 1.65 in addition to CR criteria is required.
  • sCR may be determined by CR + normal FLC ratio, absence of clonal plasma cells (determined by immunohistochemistry) or 2-to 4-color flow cytometry.
  • VGPR may be determined by serum, urine M-protein detectable by immunofixation, not by electrophoresis, >90% reduction, or ⁇ 100mg/24hrs (in those with only measurable disease by serum FLC levels, r >90% decrease in involved-uninvolved FLC level difference). PR may be determined by >50% reduction of serum M-protein; >90% reduction in urine M-protein or ⁇ 200 mg/24 hour, or >0% decrease in uninvolved FLC or >50% reduction in plasma cells. At baseline, >50% decrease in size of soft tissue plasmacytomas is required.
  • TTR Time to response
  • MRD negativity at a sensitivity of IO 5 is defined as the percentage of participants who have achieved MRD negative status in participants achieving suspected CR.
  • CR is defined as negative immunofixation of serum and urine, disappearance of any soft tissue plasmacytomas, and ⁇ 5 % plasma cells in bone marrow; in participants for whom only measurable disease is by serum FLC level, normal FLC ratio of 0.26 to 1.65 in addition to CR criteria is required.
  • Group 3 Percentage of Participants with MRD Negativity Status at a Sensitivity of IO 5 in Participants Achieving CR in a 5-year time frame.
  • MRD negativity at a sensitivity of IO 5 is defined as the percentage of participants who have achieved MRD negative status in participants achieving suspected CR.
  • CR is defined as negative immunofixation of serum and urine, disappearance of any soft tissue plasmacytomas, and ⁇ 5% plasma cells in bone marrow; in participants for whom only measurable disease is by serum FLC level, normal FLC ratio of 0.26 to 1.65 in addition to CR criteria is required.
  • Group 1 Duration of MRD Negativity Status at a Sensitivity of 10’ 5 in Participants Achieving MRD Negativity up to approximately 2 years after treatment.
  • Duration of MRD negativity (IO -5 ) is defined as the duration from the start of MRD negative status to the time of reappearance of detectable MRD, PD, or death.
  • Duration of MRD negativity status in Group 1 and Group 3 at a sensitivity of 10’ 5 in participants achieving MRD negativity in a 5-year time frame is defined as the duration from the start of MRD negative status to the time of reappearance of detectable MRD, PD, or death.
  • PD may be determined by increase of >25% from lowest response value in any one or more of the following: serum M-component increase >0.5 g/dL or urine M-component increase >200 mg/24-hour; difference between involved and uninvolved FLC levels increase must be >10 mg/dL; bone marrow plasma cell >10%; definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas; or development of hypercalcemia that can be attributed solely to plasma cell proliferative disorder.
  • Example 2 Synergistic effect in tumor inhibition with a combination of CD38-binding fusion protein, immunomodulatory drugs and proteasome inhibitors in a mouse model
  • NCI-H929 is a differentiated, highly secretory human plasma cell line established from a malignant effusion obtained from a patient with IgAK myeloma.
  • Group 1 received the PBS vehicle twice weekly (BIW) for three weeks.
  • Group 2 received the CD38-binding fusion protein at 2.5 mg/kg BIW for three weeks.
  • Group 3 received lenalidomide at 25 mg/kg, BIW for 3 weeks, or bortezomib at 1.0 mg/kg, BIW for 3 weeks.
  • Group 4 received a combination of a CD38-binding fusion protein and lenalidomide, or the combination of a CD38-binding fusion protein and bortezomib (at the same dosing amounts and schedules as the single doses).
  • mice models The treatment effects on mice models are demonstrated in FIG. 1A and FIG. IB.
  • the results show that more significant reduction in tumor median volume was achieved in groups treated with a combination therapy, compared to groups treated with the CD38-binding fusion protein or lenalidomide or bortezomib alone.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Physics & Mathematics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Plant Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des méthodes de traitement du cancer (par exemple, un cancer exprimant CD38 tel qu'un myélome multiple) à l'aide d'une protéine de fusion se liant à CD38 comprenant un anticorps anti-CD38 fusionné à une ou plusieurs protéines d'interféron alpha-2b atténuées en combinaison avec un ou plusieurs (par exemple, un, deux ou plus) agents pour traiter le cancer. Dans certains modes de réalisation, le cancer est un myélome multiple. Dans certains modes de réalisation, lesdits un ou plusieurs agents comprennent un médicament immunomodulateur (par exemple, le lénalidomide et/ou le pomalidomide) et/ou des inhibiteurs de protéasome (par exemple, le bortézomib et/ou le carfilzomib).
PCT/IB2023/000653 2022-09-29 2023-09-28 Thérapie de combinaison de protéines de fusion se liant à cd38 WO2024069240A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263411286P 2022-09-29 2022-09-29
US63/411,286 2022-09-29

Publications (2)

Publication Number Publication Date
WO2024069240A2 true WO2024069240A2 (fr) 2024-04-04
WO2024069240A3 WO2024069240A3 (fr) 2024-05-23

Family

ID=89378574

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/000653 WO2024069240A2 (fr) 2022-09-29 2023-09-28 Thérapie de combinaison de protéines de fusion se liant à cd38

Country Status (1)

Country Link
WO (1) WO2024069240A2 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6713446B2 (en) 2001-01-25 2004-03-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Formulation of boronic acid compounds
US7232818B2 (en) 2004-04-15 2007-06-19 Proteolix, Inc. Compounds for enzyme inhibition
US10544199B2 (en) 2014-10-29 2020-01-28 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101912957B1 (ko) * 2010-09-27 2018-10-29 모르포시스 아게 다발성 골수종 및 nhl 치료를 위한 항-cd38 항체 및 레날리도마이드 또는 보르테조밉
UA119352C2 (uk) * 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Комбінація леналідоміду або помалідоміду і конструкції анти-cd38 антитіло-атенуйований інтерферон альфа-2b та спосіб лікування суб'єкта, який має cd38-експресуючу пухлину

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6713446B2 (en) 2001-01-25 2004-03-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Formulation of boronic acid compounds
US6958319B2 (en) 2001-01-25 2005-10-25 The United States Of America As Represented By The Department Of Health And Human Services Formulation of boronic acid compounds
US7232818B2 (en) 2004-04-15 2007-06-19 Proteolix, Inc. Compounds for enzyme inhibition
US10544199B2 (en) 2014-10-29 2020-01-28 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants
US11319356B2 (en) 2014-10-29 2022-05-03 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants

Also Published As

Publication number Publication date
WO2024069240A3 (fr) 2024-05-23

Similar Documents

Publication Publication Date Title
JP6767029B2 (ja) 疾患に対する免疫反応を誘導するための併用療法
TWI754622B (zh) Ctla4結合劑
US11597768B2 (en) Immunotherapy for hepatocellular carcinoma
KR20160146770A (ko) 레날리도마이드 또는 포말리도마이드 및 cd38 항체-감쇠 인터페론-알파 구성체의 조합, 및 이의 용도
JP2022169709A (ja) 関節リウマチの治療
WO2015126548A1 (fr) Traitement d'une maladie par induction d'une réponse immune à des cellules exprimant le trop-2
EP4173636A1 (fr) Utilisation combinée d'un anticorps anti-ccr8 et d'un agent chimiothérapeutique
AU2020208193A1 (en) Methods of treating cancer with a PD-1 axis binding antagonist and an RNA vaccine
US20230272041A1 (en) Formulation, Dosage Regimen, and Manufacturing Process for Heterodimeric FC-Fused Proteins
JP2015517511A (ja) Cd37抗体とice(イフォスファミド、カルボプラチン、エトポシド)の併用
JP2023542257A (ja) 癌の治療のために二重特異性t細胞誘導分子の治療用量を投与する方法
US20230250176A1 (en) Ppharmaceutical formulations and therapeutic uses of multi-specific binding proteins that bind egfr, nkg2d, and cd16
WO2024069240A2 (fr) Thérapie de combinaison de protéines de fusion se liant à cd38
WO2023146394A1 (fr) Polythérapie pour le traitement du cancer
WO2022240688A1 (fr) Schéma posologique pour polythérapie ciblant dll3 et pd-1
US20210130460A1 (en) Anti-cd19 therapy in combination with lenalidomide for the treatment of leukemia or lymphoma
JP2023501375A (ja) 抗dll3剤の投与レジメン
WO2024073716A2 (fr) Thérapie combinatoire de protéines de fusion se liant à cd38
TW202410916A (zh) Cd38結合融合蛋白之給藥
WO2023227949A1 (fr) Dosage de protéine de fusion se liant à cd38
JP2016527200A (ja) Cd37抗体とクロラムブシルの併用
JP7132256B2 (ja) 関節リウマチの治療
WO2024041652A1 (fr) Méthodes de traitement du cancer
WO2023001118A1 (fr) Application d'anticorps anti-ox40 dans un médicament combiné
TW202332698A (zh) 癌症之組合療法

Legal Events

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

Ref document number: 23828792

Country of ref document: EP

Kind code of ref document: A2