Docket No.258199.062002 (JBI6858WOPCT1) METHODS FOR TREATING MULTIPLE MYELOMA CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to United States Provisional Application Serial Number 63/595,095, filed November 1, 2023, and United States Provisional Application Serial Number 63/644,276, filed May 8, 2024, the entire contents of which are incorporated herein by reference in their entirety. REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY [0002] This application contains a sequence listing, which is submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on October 4, 2024, is named “258199062002 (JBI6858WOPCT1) Sequence Listing.xml” and is 26,313 bytes in size. FIELD OF THE INVENTION [0003] Methods of treating multiple myeloma are disclosed. BACKGROUND OF THE INVENTION [0004] Multiple myeloma (MM) is a cancer of the plasma cells. Mechanistically, multiple myeloma is characterized by production of monoclonal proteins (M-proteins) comprised of pathological immunoglobulins or fragments of such, which have lost their function. The proliferation of multiple myeloma cells leads to subsequent displacement from the normal bone marrow niche, while overproduction of M-proteins causes characteristic osteolytic lesions, increased susceptibility to infections, hypercalcemia, renal insufficiency or failure, and neurological complications. [0005] Treatment options for multiple myeloma have improved over time and vary depending on the aggressiveness of the disease, underlying prognostic factors, physical condition of the patient, and existing comorbidities. Therapeutic options include proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), alkylating agents, monoclonal antibodies (mAbs), antibody drug conjugate, histone deacetylase inhibitor, nuclear protein export inhibitor, chimeric antigen receptor (CAR) T cell therapy and stem cell transplantation. [0006] Despite these therapeutic achievements, the disease recurs and is associated with additional risk factors (e.g., comorbidities or increasing age), thus warranting the need for novel therapeutic approaches, such as new dosage and treatment regimens. In newly
Docket No.258199.062002 (JBI6858WOPCT1) diagnosed patients for whom stem cell transplantation is not a viable option, such as elderly patients, and in patients with refractory disease who have exhausted standard-of-care therapies, multiple myeloma remains an incurable malignancy and an unmet medical need with significant morbidity and mortality. There remains a need for therapeutic regimens that achieve rapid, deep and durable clinical responses, while providing manageable safety profiles. SUMMARY OF THE INVENTION [0007] According to particular embodiments of the present invention, monthly (Q4W) dosing of a BCMAxCD3 bispecific antibody (e.g., teclistamab) begins in the first 28-day BCMAxCD3 treatment cycle following a 28-day step-up phase in which one or more step-up doses (and optionally one or more treatment doses) of the BCMAxCD3 bispecific antibody are administered. [0008] An embodiment of the present invention provides a method of treating multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein: one or more step-up doses of a BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject during a step-up phase, and treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. [0009] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a BCMA binding domain comprising the HCDR1 of SEQ ID NO: 4, the HCDR2 of SEQ ID NO: 5, the HCDR3 of SEQ ID NO: 6, the LCDR1 of SEQ ID NO: 7, the LCDR2 of SEQ ID NO: 8 and the LCDR3 of SEQ ID NO: 9, and a CD3 binding domain comprising the HCDR1 of SEQ ID NO: 14, the HCDR2 of SEQ ID NO: 15, the HCDR3 of SEQ ID NO: 16, the LCDR1 of SEQ ID NO: 17, the LCDR2 of SEQ ID NO: 18 and the LCDR3 of SEQ ID NO: 19. [0010] In certain embodiments, the BCMA binding domain comprises a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 11, and the CD3 binding domain comprises a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 20 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 21.
Docket No.258199.062002 (JBI6858WOPCT1) [0011] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG1, an IgG2, an IgG3 or an IgG4 isotype. [0012] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype. [0013] In certain embodiments, the BCMAxCD3 bispecific antibody comprises one or more substitutions in its Fc region. [0014] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises S228P, F234A and L235A substitutions in its Fc region (according to EU numbering). [0015] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises S228P, F234A, L235A F405L and R409K substitutions in its Fc region (according to EU numbering). [0016] In certain embodiments, the Fc region of the BCMA-binding arm comprises S228P, F234A and L235A substitutions in its Fc region (according to EU numbering). [0017] In certain embodiments, the Fc region of the CD3-binding arm comprises S228P, F234A, L235A, F405L, and R409K substitutions in its Fc region (according to EU numbering). [0018] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 23. [0019] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 23. [0020] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 95% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 95% identity to the amino acid sequence of SEQ ID NO: 23.
Docket No.258199.062002 (JBI6858WOPCT1) [0021] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 98% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 98% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 23. [0022] In certain embodiments, the BCMAxCD3 bispecific antibody is teclistamab. [0023] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg or 3 mg/kg. [0024] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg. [0025] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 3 mg/kg. [0026] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody administered to the subject on the monthly dosing schedule (Q4W) is administered on Day 1 of each BCMAxCD3 treatment cycle. [0027] In certain embodiments, the regimen does not include any BCMAxCD3 treatment cycles in which the BCMAxCD3 bispecific antibody is administered to the subject on a bi- weekly dosing schedule (Q2W). [0028] In certain embodiments, the step-up phase is 28 days. [0029] In certain embodiments, 1-3 step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase. [0030] In certain embodiments, two step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase. [0031] In certain embodiments, the step-up phase comprises a first step-up dose of 0.06 mg/kg of the BCMAxCD3 bispecific antibody. [0032] In certain embodiments, the step-up phase comprises a first step-up dose of 0.06 mg/kg of the BCMAxCD3 bispecific antibody and a second step-up dose of 0.3 mg/kg of the BCMAxCD3 bispecific antibody.
Docket No.258199.062002 (JBI6858WOPCT1) [0033] In certain embodiments, one or more treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0034] In certain embodiments, two treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0035] In certain embodiments, three treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0036] In certain embodiments, the step-up phase comprises subcutaneously administering two step-up doses and two treatment doses of the BCMAxCD3 bispecific antibody. [0037] In certain embodiments, the step-up phase comprises subcutaneously administering two step-up doses and three treatment doses of the BCMAxCD3 bispecific antibody. [0038] In certain embodiments, the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 0.72 mg/kg or 1.5 mg/kg or 3.0 mg/kg. [0039] In certain embodiments, each of the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 1.5 mg/kg. [0040] In certain embodiments, two or three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). [0041] In certain embodiments, two treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). [0042] In certain embodiments, three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). [0043] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg and a second treatment dose of 1.5 mg/kg. [0044] In certain embodiments, the second step-up dose is administered 2-4 days after first step-up dose. [0045] In certain embodiments, the second step-up dose is administered 2 days after first step-up dose.
Docket No.258199.062002 (JBI6858WOPCT1) [0046] In certain embodiments, the first treatment dose is administered 4-7 days after the second step-up dose. [0047] In certain embodiments, the first treatment dose is administered 5 days after the second step-up dose. [0048] In certain embodiments, the second treatment dose is administered 5-9 days after the first treatment dose. [0049] In certain embodiments, the second treatment dose is administered 7 days after the first treatment dose. [0050] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15. [0051] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg, a second treatment dose of 1.5 mg/kg and a third treatment dose of 1.5 mg/kg. [0052] In certain embodiments, the third treatment dose is administered 5-9 days after the second treatment dose. [0053] In certain embodiments, the third treatment dose is administered 7 days after the second treatment dose. [0054] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22. [0055] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0056] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22; and each subsequent
Docket No.258199.062002 (JBI6858WOPCT1) BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0057] In certain embodiments, the treatment doses of the BCMAxCD3 bispecific antibody are administered to the subject on the monthly dosing schedule (Q4W) regardless of whether, or to what extent, the subject exhibits a clinical response to the regimen. [0058] In certain embodiments, the step-up phase is in Cycle 1 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 2 of the regimen. [0059] In certain embodiments, the regimen is a combination therapy comprising administration of other anti-multiple myeloma agents in addition to the BCMAxCD3 bispecific antibody. [0060] In certain embodiments, the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen, or later. [0061] In certain embodiments, the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen. [0062] In certain embodiments, during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg and a fourth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. [0063] In certain embodiments, the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose and the fourth dose are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter. [0064] In certain embodiments, during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg, a fourth dose of 1.5 mg/kg and a fifth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase.
Docket No.258199.062002 (JBI6858WOPCT1) [0065] In certain embodiments, the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter. [0066] In certain embodiments, the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose and the fourth dose of the BCMAxCD3 bispecific antibody are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 2, respectively, and the treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. [0067] In certain embodiments, the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. [0068] In certain embodiments, the regimen is a combination therapy comprising administration of the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab and lenalidomide. In certain embodiments, administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3. [0069] All methods described herein, however expressed, may be described as corresponding uses, in particular medical uses. DETAILED DESCRIPTION OF THE INVENTION [0070] The disclosed methods can be understood more readily by reference to the following detailed description. It is to be understood that the disclosed methods are not limited to the specific methods described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is
Docket No.258199.062002 (JBI6858WOPCT1) not intended to be limiting of the claimed methods. All patents, published patent applications and publications cited herein are incorporated by reference as if set fourth fully herein. [0071] As used herein, the singular forms “a,” “an,” and “the” include the plural. [0072] Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein. [0073] “About” when used in reference to numerical ranges, cutoffs, or specific values means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. Unless explicitly stated otherwise within the Examples or elsewhere in the Specification in the context of an assay, result or embodiment, “about” means within one standard deviation per the practice in the art, or a range of up to 5%, whichever is larger. [0074] “Antibodies” is meant in a broad sense and includes immunoglobulin molecules including monoclonal antibodies including murine, human, humanized and chimeric monoclonal antibodies, antigen binding fragments, multispecific antibodies, such as bispecific, trispecific, tetraspecific etc., dimeric, tetrameric or multimeric antibodies, single chain antibodies, domain antibodies and any other modified configuration of the immunoglobulin molecule that comprises an antigen binding site of the required specificity. “Full length antibodies” are comprised of two heavy chains (HC) and two light chains (LC) inter-connected by disulfide bonds as well as multimers thereof (e.g. IgM). Each heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region (comprised of domains CH1, hinge, CH2 and CH3). Each light chain is comprised of a light chain variable region (VL) and a light chain constant region (CL). The VH and the VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with framework regions (FR). Each VH and VL is composed of three CDRs and four FR segments, arranged from amino-to-carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. Immunoglobulins can be assigned to five major classes, IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified as the isotypes IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4. Antibody light chains of any vertebrate species can be assigned to one of two clearly distinct types, namely kappa (κ) and lambda (λ), based on the amino acid sequences of their constant domains.
Docket No.258199.062002 (JBI6858WOPCT1) [0075] “Antigen binding fragment” or “antigen binding domain” refers to a portion of an immunoglobulin molecule that binds an antigen. Antigen binding fragments can be synthetic, enzymatically obtainable or genetically engineered polypeptides and include the VH, the VL, the VH and the VL, Fab, F(ab')2, Fd and Fv fragments, domain antibodies (dAb) consisting of one VH domain or one VL domain, shark variable IgNAR domains, camelized VH domains, minimal recognition units consisting of the amino acid residues that mimic the CDRs of an antibody, such as FR3-CDR3-FR4 portions, the HCDR1, the HCDR2 and/or the HCDR3 and the LCDR1, the LCDR2 and/or the LCDR3. VH and VL domains can be linked together via a synthetic linker to form various types of single chain antibody designs where the VH/VL domains can pair intramolecularly, or intermolecularly in those cases when the VH and VL domains are expressed by separate single chain antibody constructs, to form a monovalent antigen binding site, such as single chain Fv (scFv) or diabody; described for example in Int. Patent Publ. Nos. WO1998/44001, WO1988/01649, WO1994/13804 and WO1992/01047. [0076] “BCMA” refers to human B-cell maturation antigen, also known as CD269 or TNFRSF17 (UniProt Q02223). The extracellular domain of BCMA encompasses residues 1- 54 of Q02223. Human BCMA comprises the amino acid sequence of SEQ ID NO: 1. SEQ ID NO: 1 MLQMAGQCSQNEYFDSLLHACIPCQLRCSSNTPPLTCQRYCNASVTNSVKGT NAILWTCLGLSLIISLAVFVLMFLLRKINSEPLKDEFKNTGSGLLGMANIDLEK SRTGDEIILPRGLEYTVEECTCEDCIKSKPKVDSDHCFPLPAMEEGATILVTTK TNDYCKSLPAALSATEIEKSISAR [0077] “Bispecific” refers to an antibody that specifically binds two distinct antigens or two distinct epitopes within the same antigen. The bispecific antibody can have cross- reactivity to other related antigens, for example to the same antigen from other species (homologs), such as human or monkey, for example Macaca cynomolgus (cynomolgus, cyno) or Pan troglodytes, or can bind an epitope that is shared between two or more distinct antigens. [0078] “BCMAxCD3 bispecific antibody” refers to a bispecific antibody that specifically binds BCMA and CD3. [0079] “Cancer” refers to a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth results in the formation of malignant tumors that invade neighboring tissues and can also
Docket No.258199.062002 (JBI6858WOPCT1) metastasize to distant parts of the body through the lymphatic system or bloodstream. A “cancer” or “cancer tissue” can include a tumor. [0080] “CD3” refers to a human antigen which is expressed on T cells as part of the multimolecular T cell receptor (TCR) complex and which consists of a homodimer or heterodimer formed from the association of two or four receptor chains: CD3 epsilon, CD3 delta, CD3 zeta and CD3 gamma. Human CD3 epsilon comprises the amino acid sequence of SEQ ID NO: 2. SEQ ID NO: 3 shows the extracellular domain of CD3 epsilon. SEQ ID NO: 2 MQSGTHWRVLGLCLLSVGVWGQDGNEEMGGITQTPYKVSISGTTVILTCPQY PGSEILWQHNDKNIGGDEDDKNIGSDEDHLSLKEFSELEQSGYYVCYPRGSKP EDANFYLYLRARVCENCMEMDVMSVATIVIVDICITGGLLLLVYYWSKNRK AKAKPVTRGAGAGGRQRGQNKERPPPVPNPDYEPIRKGQRDLYSGLNQRRI SEQ ID NO: 3 DGNEEMGGITQTPYKVSISGTTVILTCPQYPGSEILWQHNDKNIGGDEDDKNI GSDEDHLSLKEFSELEQSGYYVCYPRGSKPEDANFYLYLRARVCENCMEMD [0081] “CH3 region” or “CH3 domain” refers to the CH3 region of an immunoglobulin. The CH3 region of human IgG1 antibody corresponds to amino acid residues 341-446. However, the CH3 region can also be any of the other antibody isotypes as described herein. [0082] “Combination” means that two or more therapeutics are administered to a subject. As used herein, a “combination therapy” refers to a therapeutically effective regimen that comprises administration of two or more anti-multiple myeloma agents to a subject to treat multiple myeloma; for example, a combination therapy may comprise administration of daratumumab and/or lenalidomide, in addition to teclistamab. [0083] “Complementarity determining regions” (CDR) are antibody regions that bind an antigen. CDRs can be defined using various delineations such as Kabat (Wu et al. J Exp Med 132: 211-50, 1970) (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991), Chothia (Chothia et al. J Mol Biol 196: 901-17, 1987), IMGT (Lefranc et al. Dev Comp Immunol 27: 55-77, 2003) and AbM (Martin and Thornton J Bmol Biol 263: 800-15, 1996). The correspondence between the various delineations and variable region numbering are described (see e.g. Lefranc et al. Dev Comp Immunol 27: 55-77, 2003; Honegger and Pluckthun, J Mol Biol 309:657-70, 2001; International ImMunoGeneTics (IMGT) database; Web resources, http://www_imgt_org). Available programs such as abYsis by UCL Business
Docket No.258199.062002 (JBI6858WOPCT1) PLC can be used to delineate CDRs. The term “CDR”, “HCDR1”, “HCDR2”, “HCDR3”, “LCDR1”, “LCDR2” and “LCDR3” as used herein includes CDRs defined by any of the methods described supra, Kabat, Chothia, IMGT or AbM, unless otherwise explicitly stated in the specification. Preferably, the term “CDR”, “HCDR1”, “HCDR2”, “HCDR3”, “LCDR1”, “LCDR2” and “LCDR3” as used herein includes CDRs defined by the method of Kabat. [0084] “Comprising” is intended to include examples encompassed by the terms “consisting essentially of” and “consisting of”; similarly, the term “consisting essentially of” is intended to include examples encompassed by the term “consisting of.” Unless the context clearly requires otherwise, throughout the description and the claims, the words “comprise”, “comprising”, and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of “including, but not limited to”. [0085] “Fc gamma receptor” (FcJR) refers to well-known FcJRI, FcJRIIa, FcJRIIb or FcJRIII. Activating FcJR includes FcJRI, FcJRIIa and FcJRIII. [0086] “Human antibody” refers to an antibody that is optimized to have minimal immune response when administered to a human subject. Variable regions of human antibody are derived from human immunoglobulin sequences. If human antibody contains a constant region or a portion of the constant region, the constant region is also derived from human immunoglobulin sequences. Human antibody comprises heavy and light chain variable regions that are “derived from” sequences of human origin if the variable regions of the human antibody are obtained from a system that uses human germline immunoglobulin or rearranged immunoglobulin genes. Such exemplary systems are human immunoglobulin gene libraries displayed on phage, and transgenic non-human animals such as mice or rats carrying human immunoglobulin loci. “Human antibody” typically contains amino acid differences when compared to the immunoglobulins expressed in humans due to differences between the systems used to obtain the human antibody and human immunoglobulin loci, introduction of somatic mutations or intentional introduction of substitutions into the frameworks or CDRs, or both. Typically, “human antibody” is at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical in amino acid sequence to an amino acid sequence encoded by human germline immunoglobulin or rearranged immunoglobulin genes. In some cases, “human antibody” can contain consensus framework sequences derived from human framework sequence analyses, for example as described in Knappik et al., (2000) J Mol Biol 296:57-86, or synthetic
Docket No.258199.062002 (JBI6858WOPCT1) HCDR3 incorporated into human immunoglobulin gene libraries displayed on phage, for example as described in Shi et al., (2010) J Mol Biol 397:385-96, and in Int. Patent Publ. No. WO2009/085462. Antibodies in which at least one CDR is derived from a non-human species are not included in the definition of “human antibody”. [0087] “Humanized antibody” refers to an antibody in which at least one CDR is derived from non-human species and at least one framework is derived from human immunoglobulin sequences. Humanized antibody can include substitutions in the frameworks so that the frameworks can not be exact copies of expressed human immunoglobulin or human immunoglobulin germline gene sequences. [0088] “Identity” refers to a relationship between the sequences of two or more polypeptide molecules or two or more nucleic acid molecules, as determined by aligning and comparing the sequences. “Percent (%) sequence identity” with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, or MEGALIGN (DNAStar, Inc.) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. [0089] “Isolated” refers to a homogenous population of molecules (such as synthetic polynucleotides or a protein such as an antibody) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step. “Isolated antibody” refers to an antibody that is substantially free of other cellular material and/or chemicals and encompasses antibodies that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity. [0090] “Monoclonal antibody” refers to an antibody obtained from a substantially homogenous population of antibody molecules, i.e., the individual antibodies comprising the population are identical except for possible well-known alterations such as removal of C- terminal lysine from the antibody heavy chain or post-translational modifications such as
Docket No.258199.062002 (JBI6858WOPCT1) amino acid isomerization or deamidation, methionine oxidation or asparagine or glutamine deamidation. Monoclonal antibodies typically bind one antigenic epitope. A bispecific monoclonal antibody binds two distinct antigenic epitopes. Monoclonal antibodies can have heterogeneous glycosylation within the antibody population. Monoclonal antibody can be monospecific or multispecific such as bispecific, monovalent, bivalent or multivalent. [0091] “Mutation” refers to an engineered or naturally occurring alteration in a polypeptide or polynucleotide sequence when compared to a reference sequence. The alteration can be a substitution, insertion or deletion of one or more amino acids or polynucleotides. [0092] “Negative minimal residual disease status” or “negative MRD status” or “MRD negative” refers to the PerMillionCount (i.e., a point estimate of malignant myeloma cells per million nucleated cells) in a patients on-study bone marrow sample relative to their reference bone marrow sample (i.e., Teclistamab treatment naïve bone marrow sample). Based on this PerMillionCount, each sample is determined to be positive or negative. Samples are positive if the PerMillionCount is greater than or equal to the limit of sensitivity, otherwise they are negative. Negative minimal residual disease status can be determined at a sensitivity of 0.01% (10
-4), 0.001% (10
-5) or 0.0001% (10
-6). Negative minimal residual disease status was determined using next generation sequencing (NGS). [0093] “Pharmaceutical composition” refers to composition that comprises an active ingredient and a pharmaceutically acceptable carrier. [0094] “Pharmaceutically acceptable carrier” or “excipient” refers to an ingredient in a pharmaceutical composition, other than the active ingredient, which is nontoxic to a subject. [0095] “Recombinant” refers to DNA, antibodies and other proteins that are prepared, expressed, created or isolated by recombinant means when segments from different sources are joined to produce recombinant DNA, antibodies or proteins. [0096] “Refractory” refers to a cancer that is not amendable to surgical intervention and is initially unresponsive to therapy. [0097] “Relapsed” refers to a cancer that responded to treatment but then returns. [0098] “Step-up dose” refers to a dose of an active agent that is administered to a subject prior to a treatment dose. A step-up dose is lower than the treatment dose. To prevent or lessen certain toxicities, such as cytokine release syndrome (CRS), a “priming” dose strategy may include one or more lower step-up dose(s) followed by higher treatment doses. As used herein with respect to a BCMAxCD3 bispecific antibody, a “step-up phase” refers to an initial phase of a therapeutically effective regimen in which at least one step-up dose of a
Docket No.258199.062002 (JBI6858WOPCT1) BCMAxCD3 bispecific antibody is administered to the subject. A step-up phase may also include one or more treatment doses, i.e., a step-up phase may include one or more step-up doses followed by one or more treatment doses; for example, a step-up phase may include two step-up doses followed by two treatment doses. In preferred embodiments, the step-up phase is 28 days, i.e., the step-up phase is a 28-day cycle of a therapeutically effective regimen. [0099] “Subject” as used herein refers to a human subject. Except when noted, the terms “patient” and “subject” are used interchangeably. [0100] “T cell redirecting therapeutic” refers to a molecule containing two or more binding regions, wherein one of the binding regions specifically binds a cell surface antigen on a target cell or tissue and wherein a second binding region of the molecule specifically binds a T cell antigen. Examples of cell surface antigen include a tumor associated antigen, such as BCMA. Examples of T cell antigen include, e.g., CD3. This dual/multi-target binding ability recruits T cells to the target cell or tissue leading to the eradication of the target cell or tissue. [0101] “Therapeutically effective amount” refers to an amount effective, at doses and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount can vary depending on factors such as the disease state, age, sex, and weight of the individual, and the ability of a therapeutic or a combination of therapeutics to elicit a desired response in the individual. Exemplary indicators of an effective therapeutic or combination of therapeutics that include, for example, improved well-being of the patient. [0102] “Treat” or “treatment” refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder. Beneficial or desired clinical results include alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if a subject was not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented. [0103] “Treatment dose” refers to a dose of the active agent that is administered to a subject to treat a disease. A treatment dose may be administered at a regular dosing interval on a repetitive basis (e.g. weekly, biweekly, monthly). A treatment dose may be preceded by
Docket No.258199.062002 (JBI6858WOPCT1) one or more step-up doses. In certain embodiments, a dosing regimen may include treatment doses in different amounts, e.g., one or more treatment doses of 1.5 mg/kg, one or more treatment doses of 3 mg/kg, etc. [0104] A patient that is “triple-class exposed” refers to a multiple myeloma patient that has previously been treated with (at a minimum) a proteasome inhibitor, an immunomodulatory agent and an anti-CD38 monoclonal antibody. [0105] “Tumor cell” or a “cancer cell” refers to a cancerous, pre-cancerous or transformed cell, either in vivo, ex vivo, or in tissue culture, that has spontaneous or induced phenotypic changes. These changes do not necessarily involve the uptake of new genetic material. Although transformation can arise from infection with a transforming virus and incorporation of new genomic nucleic acid, uptake of exogenous nucleic acid or it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene. Transformation/cancer is exemplified by morphological changes, immortalization of cells, aberrant growth control, foci formation, proliferation, malignancy, modulation of tumor specific marker levels, invasiveness, tumor growth in suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo. [0106] The numbering of amino acid residues in the antibody constant region throughout the specification is according to the EU index as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991), unless otherwise explicitly stated. Antibody constant chain numbering can be found for example at ImMunoGeneTics website, at IMGT Web resources at IMGT Scientific charts. [0107] Conventional one and three-letter amino acid codes are used herein as shown in Table 1. Table 1. Amino acid Three-letter code One-letter code Alanine Ala A Arginine Arg R Asparagine Asn N Aspartate Asp D Cysteine Cys C Glutamate Gln E Glutamine Glu Q
Docket No.258199.062002 (JBI6858WOPCT1) Glycine Gly G Histidine His H Isoleucine Ile I Leucine Leu L Lysine Lys K Methionine Met M Phenylalanine Phe F Proline Pro P Serine Ser S Threonine Thr T Tryptophan Trp W Tyrosine Tyr Y Valine Val V BCMAxCD3 bispecific antibodies and uses thereof [0108] It is well-known in the art that drug development is an unpredictable field. The lack of predictability in the art is evidenced, for example, by health authority requirements (such as those of the Food and Drug Administration) to establish a safe and effective dosing regimen for each individual drug candidate in clinical trials. Over the last decade (2011- 2020), only 7.9% of all developmental drug candidates achieved FDA approval from a Phase I clinical study. See Clinical Development Success Rates and Contributing Factors 2011- 2020. The rate of success is even lower in oncology, such that only 5.3% of oncology drug candidates succeed. [0109] In the field of oncology, even for a drug that already has an established dose in a particular indication, the Food and Drug Administration (FDA) recommends further clinical studies to identify an optimal dose for a new indication; otherwise, patients may be exposed to unreasonable and significant risk, among other potential deficiencies. See, e.g., Optimizing the Dosage of Human Prescription Drugs and Biological Products for the Treatment of Oncologic Diseases; Draft Guidance for Industry (January 2023). [0110] Teclistamab (also known as TECVAYLI®) is the first BCMA-directed bispecific antibody approved for the treatment of patients with relapsed or refractory multiple myeloma. Teclistamab is a bispecific B-cell maturation antigen (BCMA)-directed CD3 T-cell engager indicated for the treatment of adult patients with relapsed or refractory multiple myeloma who have received at least three or four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent and an anti-CD38 monoclonal antibody. The efficacy
Docket No.258199.062002 (JBI6858WOPCT1) of teclistamab is continuing to be evaluated in patients with relapsed or refractory multiple myeloma in a multi-center clinical study (MajesTEC-1, NCT03145181 [Phase 1] and NCT04557098 [Phase 2]). The study has included patients who had previously received at least three prior therapies, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody. In the MajesTEC-1 study, at 14.1-month median follow-up, teclistamab demonstrated rapid, deep and durable responses, with an overall response rate (ORR) of 63% and median progression-free survival (mPFS) of 11.3 months (see, e.g., Usmani SZ, et al. Lancet 2021;398:665-674, and Moreau P, et al. New Engl J Med 2022; 387: 495-505, which are incorporated by reference herein). [0111] Patients with relapsed or refractory multiple myeloma (RRMM) are already at increased risk of infection, and bispecific antibodies targeting B-cell maturation antigen (BCMA) may contribute to increased infection risk, due to on-target, off-tumor toxicity. In the MajesTEC-1 clinical study, patients (N=165) received subcutaneous teclistamab 1.5 mg/kg weekly following a step-up dosing schedule (0.06 mg/kg and 0.3 mg/kg, each separated by 2–4 days). At a median follow-up of 21.6 months (range 0.26–32.69), infections were reported in 129 patients (78.2%). Overall, grade 3/4 infections occurred in 86 patients (52.1%), most commonly pneumonia (20.6%), COVID-19 (18.8%), sepsis (6.1%), and urinary tract infection (6.1%). Grade 3/4 neutropenia occurred in 65.5% of patients. Thus, there remains a need for optimized dosing regimens of BCMAxCD3 bispecific antibodies with improved safety profiles. [0112] In patients with newly diagnosed multiple myeloma, as well as patients with relapsed or refractory disease who have exhausted several therapies, multiple myeloma remains an incurable malignancy and an unmet medical need with significant morbidity and mortality. Based, in part, on evolving data in the teclistamab clinical development program (e.g., clinical data across multiple dosing cohorts and population PK models based on such clinical data) the present inventors developed modified dosing schedules that may provide improved safety profiles and strong efficacy. Such modified dosing schedules may vary, depending on the specific indication or patient population (e.g., relapsed/refractory patients or newly diagnosed patients), and also depending on the therapy (e.g., BCMAxCD3 monotherapy) or combination of therapies administered. In accordance with certain embodiments, the inventors developed modified dosing schedules of teclistamab that comprise monthly (Q4W) administration of teclistamab starting in its second treatment cycle (e.g., immediately following a step-up phase in Cycle 1). Embodiments of novel dosing regimens described herein for BCMAxCD3 bispecific antibodies may provide improved
Docket No.258199.062002 (JBI6858WOPCT1) safety profiles over currently approved regimens while maintaining deep and durable efficacy. Antibodies of the present invention [0113] Any suitable BCMAxCD3 bispecific antibody known to those skilled in the art in view of the present disclosure can be used in the invention. In particular embodiments, the BCMAxCD3 bispecific antibody is teclistamab. [0114] Various bispecific antibody formats include formats described herein and recombinant IgG-like dual targeting molecules, wherein the two sides of the molecule each contain the Fab fragment or part of the Fab fragment of at least two different antibodies; IgG fusion molecules, wherein full length IgG antibodies are fused to an extra Fab fragment or parts of Fab fragment; Fc fusion molecules, wherein single chain Fv molecules or stabilized diabodies are fused to heavy-chain constant-domains, Fc-regions or parts thereof; Fab fusion molecules, wherein different Fab-fragments are fused together; ScFv- and diabody-based and heavy chain antibodies (e.g., domain antibodies, nanobodies) wherein different single chain Fv molecules or different diabodies or different heavy-chain antibodies (e.g. domain antibodies, nanobodies) are fused to each other or to another protein or carrier molecule, or bispecific antibodies generated by arm exchange. Exemplary bispecific formats include dual targeting molecules include Dual Targeting (DT)-Ig (GSK/Domantis), Two-in-one Antibody (Genentech) and mAb2 (F-Star), Dual Variable Domain (DVD)-Ig (Abbott), DuoBody (Genmab), Ts2Ab (MedImmune/AZ) and BsAb (Zymogenetics), HERCULES (Biogen Idec) and TvAb (Roche), ScFv/Fc Fusions (Academic Institution), SCORPION (Emergent BioSolutions/Trubion, Zymogenetics/BMS) and Dual Affinity Retargeting Technology (Fc- DART) (MacroGenics), F(ab)2 (Medarex/AMGEN), Dual-Action or Bis-Fab (Genentech), Dock-and-Lock (DNL) (ImmunoMedics), Bivalent Bispecific (Biotecnol) and Fab-Fv (UCB- Celltech), Bispecific T Cell Engager (BITE) (Micromet), Tandem Diabody (Tandab) (Affimed), Dual Affinity Retargeting Technology (DART) (MacroGenics), Single-chain Diabody (Academic), TCR-like Antibodies (AIT, ReceptorLogics), Human Serum Albumin ScFv Fusion (Merrimack) and COMBODY (Epigen Biotech), dual targeting nanobodies (Ablynx), dual targeting heavy chain only domain antibodies. Various formats of bispecific antibodies have been described, for example in Chames and Baty (2009) Curr Opin Drug Disc Dev 12: 276 and in Nunez-Prado et al., (2015) Drug Discovery Today 20(5):588-594. [0115] In some embodiments, the BCMAxCD3 bispecific antibody comprises any one of the BCMA binding domains described in WO2017/031104, the entire content of which is
Docket No.258199.062002 (JBI6858WOPCT1) incorporated herein by reference. In some embodiments, the BCMAxCD3 bispecific antibody comprises any one of the CD3 binding domains described in WO2017/031104. In some embodiments, the BCMAxCD3 bispecific antibody comprises any one of the BCMAxCD3 bispecific antibodies described in WO2017/031104. [0116] In some embodiments, the BCMAxCD3 bispecific antibody is chimeric, humanized or human. [0117] In some embodiments, the bispecific antibody is an IgG1, an IgG2, an IgG3 or an IgG4 isotype. In preferred embodiments, the bispecific antibody is an IgG4 isotype. An exemplary wild-type IgG4 comprises an amino acid sequence of SEQ ID NO: 34. SEQ ID NO: 34: ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPC PSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYV DGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPS SIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWES NGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHN HYTQKSLSLSLGK [0118] The bispecific antibody can be of any allotype. It is expected that allotype has no influence on properties of the bispecific antibodies, such as binding or Fc-mediated effector functions. Immunogenicity of therapeutic antibodies is associated with increased risk of infusion reactions and decreased duration of therapeutic response (Baert et al., (2003) N Engl J Med 348:602-08). The extent to which therapeutic antibodies induce an immune response in the host can be determined in part by the allotype of the antibody (Stickler et al., (2011) Genes and Immunity 12:213-21). Antibody allotype is related to amino acid sequence variations at specific locations in the constant region sequences of the antibody. Table 2 shows select IgG1, IgG2 and IgG4 allotypes. Table 2. Amino acid residue at position of diversity Allotype (residue numbering: EU Index) IgG2 IgG4 IgG1 189 282 309 422 214 356 358 431 G2m(n) T M
Docket No.258199.062002 (JBI6858WOPCT1) G2m(n-) P V G2m(n)/(n-) T V nG4m(a) L R G1m(17) K E M A G1m(17,1) K D L A [0119] In some embodiments, the bispecific antibody comprises one or more Fc substitutions that reduces binding of the bispecific antibody to a FcJ receptor (FcJR) and/or reduces Fc effector functions such as C1q binding, complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) or phagocytosis (ADCP). The specific substitutions can be made in comparison to the wild-type IgG4 of SEQ ID NO: 34. [0120] Fc positions that can be substituted to reduce binding of the Fc to the activating FcJR and subsequently to reduce effector function are substitutions L234A/L235A on IgG1, V234A/G237A/P238S/H268A/V309L/A330S/P331S on IgG2, F234A/L235A on IgG4, S228P/F234A/ L235A on IgG4, N297A on all Ig isotypes, V234A/G237A on IgG2, K214T/E233P/ L234V/L235A/G236-deleted/A327G/P331A/D365E/L358M on IgG1, H268Q/V309L/ A330S/P331S on IgG2, S267E/L328F on IgG1, L234F/L235E/D265A on IgG1, L234A/L235A/G237A/P238S/H268A/A330S/P331S on IgG1, S228P/F234A/L235A/G237A/P238S on IgG4, and S228P/F234A/L235A/G236- deleted/G237A/P238S on IgG4, wherein residue numbering is according to the EU index. [0121] Fc substitutions that can be used to reduce CDC are a K322A substitution. [0122] Well-known S228P substitution can further be made in IgG4 antibodies to enhance IgG4 stability. [0123] In some embodiments, the bispecific antibody comprises one or more asymmetric substitutions in a first CH3 domain or in a second CH3 domain, or in both the first CH3 domain and the second CH3 domain. [0124] In some embodiments, the one or more asymmetric substitutions is selected from the group consisting of F405L/K409R, wild-type/F405L_R409K, T366Y/F405A, T366W/F405W, F405W/Y407A, T394W/Y407T, T394S/Y407A, T366W/T394S, F405W/T394S and T366W/T366S_L368A_Y407V, L351Y_F405A_Y407V/T394W, T366I_K392M_T394W/F405A_Y407V, T366L_K392M_T394W/F405A_Y407V, L351Y_Y407A/T366A_K409F, L351Y_Y407A/T366V_K409F, Y407A/T366A_K409F and T350V_L351Y_F405A_Y407V/T350V_T366L_K392L_T394W.
Docket No.258199.062002 (JBI6858WOPCT1) [0125] In some embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises phenylalanine at position 405 and arginine at position 409 in a first heavy chain (HC1) and leucine at position 405 and lysine at position 409 in a second heavy chain (HC2), wherein residue numbering is according to the EU Index. [0126] In some embodiments, the BCMAxCD3 bispecific antibody further comprises proline at position 228, alanine at position 234 and alanine at position 235 in both the HC1 and the HC2. [0127] Tables 3 and 4 provide sequences of an exemplary embodiment of a BCMAxCD3 bispecific antibody, according to the Kabat numbering system. Table 3. Exemplary sequences of a BCMA binding arm Region Sequence SEQ ID NO: HCDR1 SGSYFWG 4 HCDR2 SIYYSGITYYNPSLKS 5 HCDR3 HDGAVAGLFDY 6 LCDR1 GGNNIGSKSVH 7 LCDR2 DDSDRPS 8 LCDR3 QVWDSSSDHVV 9 VH QLQLQESGPGLVKPSETLSLTCTVSGGSISSGSYFWGWIRQ 10 PPGKGLEWIGSIYYSGITYYNPSLKSRVTISVDTSKNQFSLK LSSVTAADTAVYYCARHDGAVAGLFDYWGQGTLVTVSS VL SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQPPG 11 QAPVVVVYDDSDRPSGIPERFSGSNSGNTATLTISRVEAGD EAVYYCQVWDSSSDHVVFGGGTKLTVL HC QLQLQESGPGLVKPSETLSLTCTVSGGSISSGSYFWGWIRQ 12 PPGKGLEWIGSIYYSGITYYNPSLKSRVTISVDTSKNQFSLK LSSVTAADTAVYYCARHDGAVAGLFDYWGQGTLVTVSS ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSW NSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYT CNVDHKPSNTKVDKRVESKYGPPCPPCPAPEAAGGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGV EVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKC
Docket No.258199.062002 (JBI6858WOPCT1) KVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQ VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDG SFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK LC SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQPPG 13 QAPVVVVYDDSDRPSGIPERFSGSNSGNTATLTISRVEAGD EAVYYCQVWDSSSDHVVFGGGTKLTVLGQPKAAPSVTLF PPSSEELQANKATLVCLISDFYPGAVTVAWKGDSSPVKAG VETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHE GSTVEKTVAPTECS Table 4. Exemplary sequences of a CD3 binding arm Region Sequence SEQ ID NO: HCDR1 TYAMN 14 HCDR2 RIRSKYNNYATYYAASVKG 15 HCDR3 HGNFGNSYVSWFAY 16 LCDR1 RSSTGAVTTSNYAN 17 LCDR2 GTNKRAP 18 LCDR3 ALWYSNLWV 19 VH EVQLVESGGGLVQPGGSLRLSCAASGFTFNTYAMNWVRQ 20 APGKGLEWVARIRSKYNNYATYYAASVKGRFTISRDDSK NSLYLQMNSLKTEDTAVYYCARHGNFGNSYVSWFAYWG QGTLVTVSS VL QTVVTQEPSLTVSPGGTVTLTCRSSTGAVTTSNYANWVQQ 21 KPGQAPRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGV QPEDEAEYYCALWYSNLWVFGGGTKLTVLGQP HC EVQLVESGGGLVQPGGSLRLSCAASGFTFNTYAMNWVRQ 22 APGKGLEWVARIRSKYNNYATYYAASVKGRFTISRDDSK NSLYLQMNSLKTEDTAVYYCARHGNFGNSYVSWFAYWG QGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDY FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPS SSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE
Docket No.258199.062002 (JBI6858WOPCT1) AAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQ FNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQD WLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPP SQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT TPPVLDSDGSFLLYSKLTVDKSRWQEGNVFSCSVMHEALH NHYTQKSLSLSLGK LC QTVVTQEPSLTVSPGGTVTLTCRSSTGAVTTSNYANWVQQ 23 KPGQAPRGLIGGTNKRAPGTPARFSGSLLGGKAALTLSGV QPEDEAEYYCALWYSNLWVFGGGTKLTVLGQPKAAPSVT LFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVK AGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVT HEGSTVEKTVAPTECS [0128] In some embodiments, the BCMAxCD3 bispecific antibody is CC-93269, BI 836909, JNJ-64007957 (teclistamab), or PF-06863135. In preferred embodiments, the BCMAxCD3 bispecific antibody is teclistamab (also referred to herein as Tec), which has the sequences described in Tables 3 and 4. [0129] Teclistamab and its methods of use are described, for example, in WO 2017/031104, WO 2019/220369 and WO 2021/228783, which are incorporated by reference herein. According to particular embodiments, the BCMAxCD3 bispecific antibody has an amino acid sequence with at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of teclistamab. [0130] Additional embodiments of BCMAxCD3 bispecific antibodies that may be used in combination regimens of the present invention are described below. [0131] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a BCMA binding domain comprising the HCDR1 of SEQ ID NO: 4, the HCDR2 of SEQ ID NO: 5, the HCDR3 of SEQ ID NO: 6, the LCDR1 of SEQ ID NO: 7, the LCDR2 of SEQ ID NO: 8 and the LCDR3 of SEQ ID NO: 9, and a CD3 binding domain comprising the HCDR1 of SEQ ID NO: 14, the HCDR2 of SEQ ID NO: 15, the HCDR3 of SEQ ID NO: 16, the LCDR1 of SEQ ID NO: 17, the LCDR2 of SEQ ID NO: 18 and the LCDR3 of SEQ ID NO: 19. [0132] In certain embodiments, the BCMA binding domain comprises a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 11, and the CD3 binding domain comprises a heavy chain variable region (VH) having the amino acid
Docket No.258199.062002 (JBI6858WOPCT1) sequence of SEQ ID NO: 20 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 21. [0133] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG1, an IgG2, an IgG3 or an IgG4 isotype. [0134] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype. [0135] In certain embodiments, the BCMAxCD3 bispecific antibody comprises one or more substitutions in its Fc region. [0136] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises S228P, F234A and L235A substitutions in its Fc region (according to EU numbering). [0137] In certain embodiments, the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises S228P, F234A, L235A F405L and R409K substitutions in its Fc region (according to EU numbering). [0138] In certain embodiments, the Fc region of the BCMA-binding arm comprises S228P, F234A and L235A substitutions in its Fc region (according to EU numbering). [0139] In certain embodiments, the Fc region of the CD3 CD3-binding arm comprises S228P, F234A, L235A, F405L, and R409K substitutions in its Fc region (according to EU numbering). [0140] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 23. [0141] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 23. [0142] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 95% identity to the amino acid
Docket No.258199.062002 (JBI6858WOPCT1) sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 95% identity to the amino acid sequence of SEQ ID NO: 23. [0143] In certain embodiments, the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 98% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 98% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 23. [0144] In certain embodiments, the BCMAxCD3 bispecific antibody is teclistamab. Patient populations with multiple myeloma [0145] The BCMAxCD3 bispecific antibodies disclosed herein are for use in treating multiple myeloma in a subject. In certain embodiments, the subject is newly diagnosed with multiple myeloma. In certain embodiments, the subject is newly diagnosed with multiple myeloma and ineligible for ASCT. In certain embodiments, the subject is newly diagnosed multiple myeloma and either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. In certain embodiments, the subject is newly diagnosed with multiple myeloma and previously received ASCT. In certain embodiments, the subject is newly diagnosed with multiple myeloma and has completed induction therapy followed by ASCT (with or without consolidation). In certain embodiments, the subject is newly diagnosed with multiple myeloma and previously received 4 to 6 cycles of 3- or 4-drug induction therapy that includes a proteasome inhibitor and/or an IMiD with or without anti- CD38 monoclonal antibody and a single or tandem ASCT (in certain embodiments, the subject has received post-ASCT consolidation for up to 2 cycles if the total number of induction plus consolidation cycles did not exceed 6). In certain embodiments, the subject is newly diagnosed with multiple myeloma and is eligible for ASCT. In certain embodiments, the subject is newly diagnosed with multiple myeloma, is eligible for ASCT, and has not yet received ASCT. Unless indicated otherwise, a diagnosis of multiple myeloma as described herein refers to a diagnosis of multiple myeloma according to IMWG 2016 criteria. [0146] In other embodiments, the subject is relapsed or refractory to treatment with one or more prior anti-cancer treatments. Relapsed disease means a cancer has come back. Refractory disease means a cancer has not improved with treatment or has stopped responding to treatment.
Docket No.258199.062002 (JBI6858WOPCT1) [0147] In some embodiments, the subject is relapsed or refractory to treatment with a therapeutic used to treat multiple myeloma or other hematological malignancies. [0148] In certain embodiments, the subject has had been treated with from 2 to 14 prior lines of therapy. [0149] In certain embodiments, the subject has received at least three prior lines of therapy. [0150] In certain embodiments, the subject has received at least four prior lines of therapy. [0151] In certain embodiments, the subject has received at least five prior lines of therapy (penta-drug exposed). [0152] In certain embodiments, the subject has received at least three prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent and an anti-CD38 monoclonal antibody. [0153] In certain embodiments, the subject has received at least four prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent and an anti-CD38 monoclonal antibody. [0154] In particular embodiments, the patients are relapsed or refractory or intolerant to the last line of therapy (LOT); were exposed to a proteasome inhibitor, immunomodulatory drug, and anti-CD38 therapy; and had measurable disease. [0155] In some embodiments, the subject has received three anti-cancer therapies prior to administration of the BCMAxCD3 bispecific antibody. In one embodiment, the three prior anti-cancer therapies are a proteasome inhibitor (PI), an immunomodulatory drug (IMiD), and an anti-CD38 antibody. In certain such embodiments, the proteasome inhibitor is bortezomib, carfilzomib or ixazomib, the immunomodulatory drug (IMiD) is lenalidomide, pomalidomide or thalidomide and the anti-CD38 antibody is daratumumab or isatuximab. [0156] In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti- CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is thalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is bortezomib, the immunomodulatory drug (IMiD) is thalidomide and the anti-CD38 antibody is isatuximab.
Docket No.258199.062002 (JBI6858WOPCT1) [0157] In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is thalidomide and the anti- CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is carfilzomib, the immunomodulatory drug (IMiD) is thalidomide and the anti-CD38 antibody is isatuximab. [0158] In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is lenalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti- CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is pomalidomide and the anti-CD38 antibody is isatuximab. In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is thalidomide and the anti-CD38 antibody is daratumumab. In one embodiment, the proteasome inhibitor is ixazomib, the immunomodulatory drug (IMiD) is thalidomide and the anti-CD38 antibody is isatuximab. [0159] In some embodiments, the subject is refractory or relapsed to treatment with one or more treatments or therapies, such as THALOMID
® (thalidomide), REVLIMID
® (lenalidomide), POMALYST
® (pomalidomide), VELCADE
® (bortezomib), NINLARO (ixazomib), KYPROLIS
® (carfilzomib), FARADYK
® (panobinostat), AREDIA
® (pamidronate), ZOMETA
® (zoledronic acid), DARZALEX
® (daratumumab), elotozumab or melphalan, Xpovio ® (Selinexor), Venclexta ® (Venetoclax), GSK 916, CAR-T therapies, or other BCMA-directed therapies. [0160] Various qualitative and/or quantitative methods can be used to determine relapse or refractory nature of the disease. Symptoms that can be associated are for example a decline or plateau of the well-being of the patient or re-establishment or worsening of various symptoms
Docket No.258199.062002 (JBI6858WOPCT1) associated with solid tumors, and/or the spread of cancerous cells in the body from one location to other organs, tissues or cells. [0161] In some embodiments, the multiple myeloma is relapsed or refractory to treatment with an anti-CD38 antibody, selinexor, venetoclax, lenalinomide, bortezomib, pomalidomide, carfilzomib, elotozumab, ixazomib, melphalan or thalidomide, or any combination thereof. [0162] In one embodiment, the anti-CD38 antibody is daratumumab. [0163] In another embodiment, the anti-CD38 antibody is isatuximab [0164] In some embodiments, the multiple myeloma is a high-risk multiple myeloma. Subjects with high-risk multiple myeloma are known to relapse early and have poor prognosis and outcome. Subjects can be classified as having high-risk multiple myeloma is they have one or more of the following cytogenetic abnormalities: t(4;14)(p16;q32), t(14;16)(q32;q23), del17p, 1qAmp, t(4;14)(p16;q32) and t(14;16)(q32;q23), t(4;14)(p16;q32) and del17p, t(14;16)(q32;q23) and del17p, or t(4;14)(p16;q32), t(14;16)(q32;q23) and del17p. In some embodiments, the subject having the high-risk multiple myeloma has one or more chromosomal abnormalities comprising: t(4;14)(p16;q32), t(14;16)(q32;q23), del17p, 1qAmp, t(4;14)(p16;q32) and t(14;16)(q32;q23), t(4;14)(p16;q32) and del17p, t(14;16)(q32;q23) and del17p; or t(4;14)(p16;q32), t(14;16)(q32;q23) and del17p, or any combination thereof. [0165] The cytogenetic abnormalities can be detected for example by fluorescent in situ hybridization (FISH). In chromosomal translocations, an oncogene is translocated to the IgH region on chromosome 14q32, resulting in dysregulation of these genes. t(4;14)(p16;q32) involves translocation of fibroblast growth factor receptor 3 (FGFR3) and multiple myeloma SET domain containing protein (MMSET) (also called WHSC1/NSD2), and t(14;16)(q32;q23) involves translocation of the MAF transcription factor C-MAF. Deletion of 17p (del17p) involves loss of the p53 gene locus. [0166] Chromosomal rearrangements can be identified using well known methods, for example fluorescent in situ hybridization, karyotyping, pulsed field gel electrophoresis, or sequencing. Monthly administration of a BCMAxCD3 bispecific antibody [0167] In accordance with certain embodiments, the inventors have developed novel dosing regimens for BCMAxCD3 bispecific antibodies that may provide improved safety profiles over currently approved regimens while maintaining deep and durable clinical responses over time. Based, in part, on evolving data in the teclistamab clinical development program (e.g.,
Docket No.258199.062002 (JBI6858WOPCT1) evolving clinical data across multiple dosing cohorts, and population PK models based on such clinical data) the present inventors developed modified dosing schedules. Such dosing schedules may vary, for example, depending on a particular indication or patient population (e.g., relapsed/refractory patients or newly diagnosed patients), and also depending on the therapy (e.g., BCMAxCD3 bispecific antibody monotherapy) or combination of therapies administered. In accordance with certain embodiments, the inventors developed modified dosing schedules of BCMAxCD3 bispecific antibodies that comprise monthly (Q4W) administration starting in the second BCMAxCD3 treatment cycle (e.g., immediately following a step-up phase in Cycle 1). According to certain embodiments, such dosing schedules do not include any BCMAxCD3 treatment cycles in which the BCMAxCD3 bispecific antibody is administered bi-weekly (Q2W). [0168] As used herein, a “BCMAxCD3 treatment cycle” refers to each treatment cycle in a therapeutically effective regimen in which at least one treatment dose of a BCMAxCD3 bispecific antibody is administered to the subject. In preferred embodiments, the first BCMAxCD3 treatment cycle in a therapeutically effective regimen is preceded by a step-up phase. According to particular embodiments, monthly (Q4W) dosing of the BCMAxCD3 bispecific antibody begins in the first BCMAxCD3 treatment cycle following a step-up phase in which one or more step-up doses (and optionally one or more treatment doses) are administered. In certain embodiments, a dosing regimen may include treatment doses in different amounts, e.g., one or more treatment doses of 1.5 mg/kg may be administered during a step-up phase and then treatment doses of 3 mg/kg may be administered in subsequent treatment cycles. [0169] “Fixed dose,” as used herein, also referred to as “flat dose,” refers to a dose that is administered to a subject without correction for the subject’s specific body weight or body surface area. A fixed dose, sometimes referred to as a flat dose, is therefore provided as an absolute amount of the agent (e.g., mg drug), and not as a weight-based amount (e.g. μg/kg or μg drug per kg body weight). For example, a subject weighing 65kg may be administered the same flat dose in milligrams as a subject weighing 85kg. A flat dose may be administered according to a pre-defined class or category of body weight, but is not modified according to the subject’s specific weight. For example, a “Flat Dose A” may be administered if a patient is greater than a pre-defined threshold weight (e.g., > 60 kg), whereas a different “Flat Dose B” may be administered if the patent is less than or equal to the pre-defined threshold weight (e.g., ≤ 60 kg).
Docket No.258199.062002 (JBI6858WOPCT1) [0170] As used herein, “weight-based” refers to administration of a dose amount that is based on the subject’s specific body weight; for example, 3 mg/kg refers to a dose of 3 milligrams of antibody per kilogram of the subject’s body weight. Unless otherwise specified herein, when a dose is described in a unit of “mg/kg” or “μg/kg,” weight-based dosing is being employed. [0171] Unless otherwise specified herein, a BCMAxCD3 bispecific antibody, such as teclistamab, is administered on a dosing schedule based on sequential 28-day treatment cycles, for example, Cycle 1 starts on Day 1 of Cycle 1 and ends on Day 28 of Cycle 1, and then Day 1 of Cycle 2 starts the day after Day 28 of Cycle 1 and ends on Day 28 of Cycle 2, and then Day 1 of Cycle 3 starts the day after Day 28 of Cycle 2 and ends on Day 28 of Cycle 3, and so on. As used herein with respect to treatment cycles, “C1” refers to Cycle 1, “C2” refers to Cycle 2, “C3” refers to Cycle 3, and so on. Multiple cycles may also be described, e.g., “C3-6” refers to Cycles 3-6 (Cycles 3, 4, 5 and 6). A cycle number with a “+” symbol refers to that cycle and all subsequent cycles, e.g., “C3+” refers to from and all

subsequent cycles (i.e., C3, C4, C5, C6, C7, and so on). [0172] According to certain embodiments, administration of the BCMAxCD3 bispecific antibody begins in Cycle 1 of a therapeutically effective regimen (e.g., Cycle 1 is a step-up phase). According to other embodiments, in which the BCMAxCD3 antibody is administered as part of a combination therapy with one or more additional anti-multiple myeloma agents, administration of the BCMAxCD3 bispecific antibody may start in Cycle 2 or later, i.e., administration of one or more other anti-multiple myeloma agents (but not the BCMAxCD3 bispecific antibody) may start in Cycle 1 and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2 or later. Thus, in accordance with such embodiments, the first treatment cycle that includes administration of the BCMAxCD3 bispecific antibody (e.g., a step-up phase) is Cycle 2 or later. [0173] As used herein “Q4W” means once every four weeks (i.e., once every 28 days), “Q2W” (also referred to as “bi-weekly” or “biweekly”) means once every two weeks, and “QW” (also referred to as “weekly”) means once weekly. Q4W is also referred to herein as “monthly.” The terms “Q4W” and “monthly” are used interchangeably to refer to once every 4 weeks or once every 28 days (e.g., in sequential 28-day cycles, a first treatment dose occurs on Day 1 of Cycle 1, a second treatment dose occurs on Day 1 of Cycle 2, etc.). Administration of a treatment dose once weekly (QW) is also referred to herein as a weekly dosing schedule; for example, a 28-day treatment cycle may have a weekly dosing schedule that comprises four doses one week apart from each other (e.g., on Days 1, 8, 15 and 22), or
Docket No.258199.062002 (JBI6858WOPCT1) three doses one week apart from each other (e.g., on Days 8, 15 and 22), or two doses one week apart from each other (e.g., on Days 8 and 15). Administration of a treatment dose once every two weeks (Q2W) is also referred to herein as a bi-weekly dosing schedule. Administration of a treatment dose once every four weeks (Q4W) is also referred to herein as a monthly dosing schedule. Dosing regimens may be described herein in terms of the dose amount and frequency; for example, “C1: 1500 μg/kg QW” refers to administration of 1500 μg/kg once per week in Cycle 1 of a therapeutically effective regimen, “C3-6: 3000 μg/kg Q2W” refers to administration of 3000 μg/kg once every two weeks from Cycle 3 through Cycle 6, “C3+: 3000 μg/kg Q4W” refers to administration of 3000 μg/kg once every four weeks starting in Cycle 3, etc. [0174] Additional abbreviations used herein include the following: CR, complete response; PR, partial response; Q2W, once every 2 weeks; Q4W, once every 4 weeks; QW, once weekly; RP2D, recommended phase 2 dose; SUD, step-up dose. [0175] As used herein, the recommended Phase 2 dose (RP2D) of teclistamab refers to the FDA-approved weight-based regimen for teclistamab monotherapy, based on the MajesTEC- 1 clinical study, which includes treatment doses of 1.5 mg/kg teclistamab subcutaneously (SC) administered weekly until disease progression or unacceptable toxicity, wherein the treatment doses are preceded by step-up doses of 0.06 and 0.3 mg/kg. [0176] As used herein, IMWG diagnostic criteria for multiple myeloma have been met when the participant was diagnosed. IMWG diagnostic criteria are known in the art. Multiple myeloma may be defined as clonal BMPCs ≥10% or biopsy-proven bony or extramedullary plasmacytoma
a and at least 1 of the following: x Evidence of end-organ damage, specifically: – C: Hypercalcemia: serum calcium >0.25 mmol/L (>1 mg/dL) higher than the ULN or >2.75 mmol/L (>11 mg/dL) – R: Renal insufficiency: creatinine clearance <40 mL per min
b or serum creatinine >177 μmol/L (>2 mg/dL) – A: Anemia: hemoglobin value of >20 g/L below the lower limit of normal, or a hemoglobin value <100 g/L – B: Bone lesions: 1 or more osteolytic lesions on skeletal radiography, CT, or PET- CT
c,d x Any 1 or more of the following biomarkers of malignancy: – Clonal BMPC%
a ≥60%
Docket No.258199.062002 (JBI6858WOPCT1) – Involved:uninvolved sFLC ratio
e ≥100 – >1 focal lesions on MRI studies
f a. Clonality should be established by showing κ/λ-light-chain restriction on flow cytometry, immunohistochemistry, or immunofluorescence. Bone marrow plasma cell percentage should preferably be estimated from a core biopsy specimen; in case of a disparity between the aspirate and core biopsy, the highest value should be used. b. Measured or estimated by validated equations. c. If bone marrow has less than 10% clonal plasma cells, more than one bone lesion is required to distinguish from solitary plasmacytoma with minimal marrow involvement. d. PET-CT=18F-fluorodeoxyglucose PET with CT. e. These values are based on the serum Freelite assay (The Binding Site Group, Birmingham, UK). The involved FLC must be ≥100 mg/L.f. Each focal lesion must be 5 mm or more in size. [0177] According to embodiments of the present invention, methods of treating multiple myeloma are effective in eliciting a clinical response in a subject as determined by International Myeloma Working Group (IMWG) response criteria. According to particular embodiments, the methods of treatment are effective in eliciting a partial response (PR), a very good partial response (VGPR), a complete response (CR) or a stringent complete response (sCR), as determined by IMWG response criteria. As used herein, overall response rate (ORR) refers to the percentage of patients in a population that achieve a partial response (PR) or better, i.e., a partial response, very good partial response, complete response or stringent complete response. IMWG criteria for response to Multiple Myeloma treatment are provided in Table A below. Table A Response Response Criteria sCR = stringent - CR as defined below, plus complete - Normal FLC ratio, and response - Absence of clonal PCs by immunohistochemistry, immunofluorescence
a or 2- to 4-color flow cytometry CR = complete - Negative immunofixation on the serum and urine, and response* - Disappearance of any soft tissue plasmacytomas, and - <5% PCs in bone marrow
Docket No.258199.062002 (JBI6858WOPCT1) - No evidence of initial M-protein isotype(s) on immunofixation of the serum and urine
b VGPR = very good - Serum and urine M-component detectable by immunofixation partial response* but not on electrophoresis, or - ≥90% reduction in serum M-protein plus urine M-protein <100 mg/24 hours - In addition to the above criteria, if present at baseline >90% reduction in the sum of the maximal perpendicular diameter (SPD) compared with baseline for soft-tissue plasmacytoma PR = partial - ≥50% reduction of serum M-protein and reduction in 24-hour response urinary M-protein by ≥90% or to <200 mg/24 hours - If the serum and urine M-protein are not measurable, a decrease of ≥50% in the difference between involved and uninvolved FLC levels is required in place of the M-protein criteria - In addition to the above criteria, if present at baseline, a ≥50% reduction in the size (SPD) of soft tissue plasmacytomas is also required. SD = Stable Not meeting criteria for sCR, CR, VGPR, PR, MR, or disease progressive disease PD = Progressive Any one or more of the following criteria: disease
c - Increase of 25% from lowest response value in any of the following 1-4: 1. Serum M-component (absolute increase must be ≥0.5 g/dL), and/or 2. Urine M-component (absolute increase must be ≥200 mg/24 hours), and/or 3. Only in participants without measurable serum and urine M- protein levels: the difference between involved and uninvolved FLC levels (absolute increase must be >10 mg/dL) 4. Only in participants without measurable serum and urine M- protein levels and without measurable disease by FLC levels, bone marrow plasma cell percentage (the absolute percentage must be ≥10%)
Docket No.258199.062002 (JBI6858WOPCT1) - Appearance of a new lesion(s), ≥50% increase from nadir in SPD of >1 lesion, or ≥50% increase in the longest diameter of a previous lesion >1 cm in short axis - Definite development of new bone lesions or definite increase in the size of existing bone lesions - ≥50% increase in circulating plasma cells (minimum of 200 cells/μL) if this is the only measure of disease CR=complete response; FLC=free light chain; IMWG=International Myeloma Working Group; M-protein=monoclonal paraprotein; MR=minimal response; PC=plasma cell; PD=progressive disease; PR=partial response; sCR=stringent complete response; SD=stable disease; VGPR=very good partial response
a Presence/absence of clonal cells is based upon the kappa/lambda ratio. An abnormal kappa/lambda ratio by immunohistochemistry or immunofluorescence requires a minimum of 100 plasma cells for analysis. An abnormal ratio reflecting presence of an abnormal clone is kappa/lambda of >4:1 or <1:2.
b In some cases, it is possible that the original M-protein light chain isotype is still detected on immunofixation but the accompanying heavy-chain component has disappeared; this would not be considered as a CR even though the heavy-chain component is not detectable, because it is possible that the clone evolved to one that secreted only light chains. Thus, if a participant has IgA lambda myeloma, then to qualify as CR there should be no IgA detectable on serum or urine immunofixation; if free lambda is detected without IgA, then it must be accompanied by a different heavy-chain isotype (IgG, IgM, etc).
c Clarifications to the criteria for coding progressive disease: bone marrow criteria for progressive disease are to be used only in participants without measurable disease by M- protein and by FLC levels; “25% increase” refers to M-protein, and FLC, and does not refer to bone lesions, or soft-tissue plasmacytomas and the “lowest response value” does not need to be a confirmed value. * Clarifications to the criteria for coding CR and VGPR in participants in whom the only measurable disease is by sFLC levels: CR in such participants indicates a normal FLC ratio of 0.26 to 1.65 in addition to CR criteria listed above. VGPR in such participants requires a ≥90% decrease in the difference between involved and uninvolved FLC levels. [0178] IMWG criteria for response to Multiple Myeloma treatment are also described, for example, in Durie et al., Kumar et al. and Rajkumar et al., which are incorporated by
Docket No.258199.062002 (JBI6858WOPCT1) reference herein: Durie BG, Harousseau JL, Miguel JS, et al. International uniform response criteria for multiple myeloma. Leukemia.2006;20(9):1467-1473; Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17(8):e328-346; Rajkumar SV, Harousseau JL, Durie B, et al. Consensus recommendations for the uniform reporting of clinical Trials: report of the International Myeloma Workshop Consensus Panel 1. Blood.2011;117(18):4691-4695. [0179] Embodiments of the present invention provide methods of treating multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential BCMAxCD3 treatment cycles (e.g., 28-day treatment cycles), wherein: one or more step-up doses of a BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject during a step-up phase, and treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. [0180] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg or 3 mg/kg. [0181] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg. [0182] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 3 mg/kg. [0183] In certain embodiments, each treatment dose of the BCMAxCD3 bispecific antibody administered to the subject on the monthly dosing schedule (Q4W) is administered on Day 1 of each BCMAxCD3 treatment cycle. [0184] In certain embodiments, the regimen does not include any BCMAxCD3 treatment cycles in which the BCMAxCD3 bispecific antibody is administered to the subject on a bi- weekly dosing schedule (Q2W). [0185] In certain embodiments, the step-up phase is 28 days. [0186] In certain embodiments, 1-3 step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase.
Docket No.258199.062002 (JBI6858WOPCT1) [0187] In certain embodiments, two step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase. [0188] In certain embodiments, the step-up phase comprises a first step-up dose of 0.06 mg/kg of the BCMAxCD3 bispecific antibody. [0189] In certain embodiments, the step-up phase comprises a first step-up dose of 0.06 mg/kg of the BCMAxCD3 bispecific antibody and a second step-up dose of 0.3 mg/kg of the BCMAxCD3 bispecific antibody. [0190] In certain embodiments, one or more treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0191] In certain embodiments, two treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0192] In certain embodiments, three treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. [0193] In certain embodiments, the step-up phase comprises subcutaneously administering two step-up doses and two treatment doses of the BCMAxCD3 bispecific antibody. [0194] In certain embodiments, the step-up phase comprises subcutaneously administering two step-up doses and three treatment doses of the BCMAxCD3 bispecific antibody. [0195] In certain embodiments, the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 0.72 mg/kg or 1.5 mg/kg or 3.0 mg/kg. [0196] In certain embodiments, each of the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 1.5 mg/kg. [0197] In certain embodiments, two or three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). [0198] In certain embodiments, two treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). [0199] In certain embodiments, three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW).
Docket No.258199.062002 (JBI6858WOPCT1) [0200] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg and a second treatment dose of 1.5 mg/kg. [0201] In certain embodiments, the second step-up dose is administered 2-4 days after first step-up dose. [0202] In certain embodiments, the second step-up dose is administered 2 days after first step-up dose. [0203] In certain embodiments, the first treatment dose is administered 4-7 days after the second step-up dose. [0204] In certain embodiments, the first treatment dose is administered 5 days after the second step-up dose. [0205] In certain embodiments, the second treatment dose is administered 5-9 days after the first treatment dose. [0206] In certain embodiments, the second treatment dose is administered 7 days after the first treatment dose. [0207] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15. [0208] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg, a second treatment dose of 1.5 mg/kg and a third treatment dose of 1.5 mg/kg. [0209] In certain embodiments, the third treatment dose is administered 5-9 days after the second treatment dose. [0210] In certain embodiments, the third treatment dose is administered 7 days after the second treatment dose. [0211] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22. [0212] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5
Docket No.258199.062002 (JBI6858WOPCT1) mg/kg on Day 15; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0213] In certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0214] In certain embodiments, the treatment doses of the BCMAxCD3 bispecific antibody are administered to the subject on the monthly dosing schedule (Q4W) regardless of whether, or to what extent, the subject exhibits a clinical response to the regimen. [0215] In certain embodiments, the step-up phase is in Cycle 1 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 2 of the regimen. [0216] In certain embodiments, the regimen is a combination therapy comprising administration of other anti-multiple myeloma agents in addition to the BCMAxCD3 bispecific antibody. [0217] In certain embodiments, the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen, or later. [0218] In certain embodiments, the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen. [0219] In certain embodiments, during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg and a fourth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. [0220] In certain embodiments, the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose and the fourth dose are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter.
Docket No.258199.062002 (JBI6858WOPCT1) [0221] In certain embodiments, during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg, a fourth dose of 1.5 mg/kg and a fifth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. [0222] In certain embodiments, the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter. [0223] In certain embodiments, the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose and the fourth dose of the BCMAxCD3 bispecific antibody are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 2, respectively, and the treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. [0224] In certain embodiments, the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. [0225] In certain embodiments, the regimen is a combination therapy comprising administration of the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab and lenalidomide. In certain embodiments, administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3.
Docket No.258199.062002 (JBI6858WOPCT1) Exemplary Embodiments: Treatment Strategies for Newly Diagnosed Multiple Myeloma [0226] In accordance with certain embodiments of the present invention, the inventors developed novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. [0227] Newly diagnosed multiple myeloma patients can be broadly categorized as either “transplant eligible” or “transplant ineligible.” Eligibility for transplant is typically based on age, fitness, and comorbidities. Systemic therapy, typically administered as a triplet or quadruplet drug regimen, is the mainstay of treatment both for transplant-eligible and transplant-ineligible patients. It is used in combination with ASCT for those eligible. DRd (daratumumab, lenalidomide and dexamethasone) or VRd (bortezomib, lenalidomide and dexamethasone) are two key treatment options for newly diagnosed transplant-ineligible patients with multiple myeloma, i.e., standard of care. [0228] In clinical practice, the treatment of newly diagnosed patients with multiple myeloma is evolving. For certain patients, high-dose therapy with ASCT is not feasible, mainly due to advanced age, comorbidities, or patient frailty. Approximately 40% of all transplant-eligible patients in the US do not receive high-dose therapy and ASCT as initial treatment. This development is driven both by the effectiveness of new induction regimens (e.g., DRd, VRd) without high-dose therapy and ASCT, as well as patient preference, and is expected to continue to increase. Recently reported and ongoing pivotal studies reflect this development and include both (1) patients who are assessed as being clinically transplant ineligible and (2) patients who are clinically transplant eligible but do not receive transplant as a first treatment option (transplant deferred). Results from several studies have shown similar Overall Survival outcomes for upfront ASCT versus use of a novel agent-based approach without ASCT, suggesting that deferral of ASCT is not associated with adverse survival outcomes and may be a viable option for appropriate candidates. The IFM2009 study showed no OS benefit for participants who were randomized to VRd + early ASCT compared with participants who received VRd + deferred ASCT until after first relapse. Current NCCN guidelines support this approach and indicate that a delayed ASCT after early stem cell collection and storage is an appropriate option (category 1 recommendation: NCCN 2023). [0229] Despite numerous therapeutic options, multiple myeloma remains incurable for the vast majority of patients. With each successive relapse, symptoms return, quality of life worsens, and duration of response typically decreases. Recent reports suggest that attrition
Docket No.258199.062002 (JBI6858WOPCT1) rates between the first and second lines of therapy are as high as 50% in patients with multiple myeloma and achieving the longest possible PFS with the first line of therapy drives OS outcomes. Therefore, there remains a significant and critical unmet need for new therapeutic options directed at alternative mechanisms of action that can better control the disease, provide deeper, more sustained responses, and yield better long-term outcomes including maintenance of health-related quality of life. [0230] In the field of oncology, even for a drug that already has an established dose in a particular indication, the Food and Drug Administration (FDA) recommends further clinical studies to identify an optimal dose for a new indication; otherwise, patients may be exposed to unreasonable and significant risk, among other potential deficiencies. See, e.g., Optimizing the Dosage of Human Prescription Drugs and Biological Products for the Treatment of Oncologic Diseases; Draft Guidance for Industry (January 2023). The present inventors have developed novel dosing regimens for BCMAxCD3 bispecific antibodies that provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy. In particular, the present inventors have developed novel dosing regimens for combination therapies, e.g., combination therapies comprising administration of teclistamab, daratumumab and lenalidomide, that may provide improved safety profiles over currently approved regimens while achieving deep and durable efficacy. [0231] Embodiments of the present invention provide novel dosing regimens for the treatment of multiple myeloma in subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. As used herein, subjects that are newly diagnosed have a diagnosis of multiple myeloma according to the IMWG diagnostic criteria and have not received any prior therapy for multiple myeloma or smoldering myeloma (but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent). Thus, newly diagnosed subjects do not have relapsed/refractory multiple myeloma (RRMM), i.e., they are not relapsed and/or refractory to prior multiple myeloma therapy (but may have received a short course of corticosteroids, not exceeding 40 mg of dexamethasone, or equivalent per day for a maximum of 4 days, total of 160 mg dexamethasone or equivalent). [0232] Subjects that are ineligible for ASCT as initial therapy can be ineligible due to (i) advanced age, or (ii) presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT. Subjects that are “not intended” for
Docket No.258199.062002 (JBI6858WOPCT1) ASCT as initial therapy are clinically fit enough to undergo ASCT but defer this treatment option. [0233] According to an embodiment, teclistamab, daratumumab and lenalidomide (“Tec- DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy. According to an embodiment, teclistamab, daratumumab and lenalidomide are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are ineligible for autologous stem cell transplant as initial therapy. [0234] According to certain embodiments, Tec-DR improves PFS (progression-free survival) and/or the rate of sustained MRD-negative CR (≥12 months) compared with DRd in subjects with newly diagnosed multiple myeloma who are ineligible or not intended for ASCT as initial therapy. [0235] According to certain embodiments, teclistamab in combination with daratumumab subcutaneous (SC) and lenalidomide provide an efficacious and safe approach with higher efficacy rates (e.g., mPFS, mOS and MRD negativity rates) compared to the existing standard of care of daratumumab, lenalidomide and dexamethasone (DRd) and/or bortezomib, lenalidomide and dexamethasone (VRd). [0236] According to alternative methods of treatment, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table B below. Table B. Alternative Treatment Method Study Cycle (each Dose Schedule Treatment cycle=28 days) Teclistamab SC Cycle 1 Step-up Dose 1 (Day 2): 0.06 mg/kg Step-up Dose 2 (Day 4): 0.3 mg/kg Treatment Dose on Days 8, 15, and 22: 1.5 mg/kg Cycle 2 Treatment dose weekly on Days 1, 8, 15, and 22: 1.5 mg/kg Cycle 3-6 Treatment dose Q2W on Days 1 and 15: 3 mg/kg
Docket No.258199.062002 (JBI6858WOPCT1) Study Cycle (each Dose Schedule Treatment cycle=28 days) Cycle 7+ Treatment dose Q4W on Day 1: 3 mg/kg Daratumumab Cycles 1 and 2 Weekly dose on Days 1, 8, 15, 22 SC (1800 mg) Cycles 3 to 6 Q2W dose on Days 1 and 15 Cycles 7+ Q4W dosing on Day 1 Lenalidomide Cycle 1 No dosing PO (25 mg) Cycles 2+ Days 1-21 from Cycle 2 onwards Dexamethasone Cycles 2 to 4 Weekly dose on Days 1, 8, 15, and 22 PO/IV (20 mg) IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous [0237] According to embodiments of the present invention provided herein, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table C below. Table C. Exemplary Treatment Method Study Treatment Dose Schedule Teclistamab SC C2: 2 step-up doses (0.06 and 0.3 mg/kg) on Days 1 and 3 followed by treatment doses (1.5 mg/kg) on Days 8 and 15 C3+: Q4W treatment doses (3 mg/kg) on Day 1 Daratumumab SC 1800 mg C1-2: weekly on Days 1, 8, 15, and 22 C3-6: Q2W on Days 1 and 15 C7+: Q4W on Day 1 Lenalidomide oral 25 mg* C1+: D1-21 Dexamethasone oral/IV 20 mg C1-2: weekly on Days 1, 8, 15, and 22 * Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl. IV=intravenous; Q2W=every 2 weeks; Q4W=every 4 weeks; SC=subcutaneous
Docket No.258199.062002 (JBI6858WOPCT1) [0238] In the regimen shown in Table C, teclistamab is administered after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete. It was believed that a DR lead-in cycle and an early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) may reduce the rate of infections compared to the regimen shown in Table B, while still achieving robust efficacy. [0239] According to an embodiment, a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, on a dosing schedule comprising sequential 28-day treatment cycles, wherein administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3. In certain embodiments, the regimen further comprises, in Cycles 1 and 2 only, orally or intravenously administering dexamethasone to the subject. [0240] According to another embodiment, a method for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy comprises: administering a therapeutically effective combination therapy to the subject comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide according to a regimen comprising 28-day cycles, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 2, subcutaneously administering a first step-up dose of 60 μg/kg (e.g., on Day 1 or Day 2), a second step-up dose of 300 μg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 μg/kg (e.g., on Days 8 and 15), and in Cycle 3 and all subsequent treatment cycles, subcutaneously administering a monthly (Q4W) dose of 3000 μg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi- weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the
Docket No.258199.062002 (JBI6858WOPCT1) lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle. [0241] In certain embodiments, the regimen further comprises, in Cycles 1-2 only, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). [0242] As discussed in the example section below, clinical data from safety run-in cohorts in a Phase 3 Teclistmab study produced unexpected findings. A Safety Run-In introduced a DRd lead-in dosing strategy (i.e., Drd dosing in Cycle 1 and introduction of Tec in Cycle 2 with step-up dosing occurring while lenalidomide is dosed). This approach was adopted to explore whether a DRd lead-in would reduce the burden of disease in newly diagnosed patients and further reduce the occurrence and severity of CRS. However, the data suggest that a lead-in cycle of DRd does not result in a reduction of the severity of CRS events. In contrast, there may be a potentially increased risk of higher-grade CRS when participants are exposed to both prior lenalidomide during Cycle 1 and concurrent lenalidomide during administration of the step-up dosing of Tec. The prior dexamethasone exposure before having started Tec may have contributed as well to the infection findings. While the DRd lead-in dosing led to higher grade CRS, early preliminary efficacy in these cohorts support the lower exposure (early Q4W dosing) of Tec. Therefore, it is believed the dosing schedule shown in Table D may minimize exposure to Tec and optimize tolerability and safety by completing step-up dosing in Cycle 1 and transitioning to monthly dosing in Cycle 2. According to certain embodiments, teclistamab and daratumumab are initiated in Cycle 1 and lenalidomide added in Cycle 2 after the step-up phase, and the less frequent (i.e., Q4W) dosing to mitigate infection is retained. According to certain embodiments, the teclistamab SC dose schedule comprises 2 step-up doses (0.06 and 0.3 mg/kg) on Days 2 and 4 of Cycle 1 followed by 1.5 mg/kg on Days 8 and 15 of Cycle 1; and starting from Cycle 2, 3 mg/kg Q4W on Day 1 of the cycle. [0243] According to certain embodiments, teclistamab, daratumumab and lenalidomide (“Tec-DR”) are administered as a combination therapy for the treatment of adult patients with newly diagnosed multiple myeloma who are either ineligible or not intended for autologous stem cell transplant as initial therapy, according to the therapeutically effective regimen shown in Table D below.
Docket No.258199.062002 (JBI6858WOPCT1) Table D. Exemplary Treatment Method Study Treatment Dose Schedule Teclistamab SC C1: 2 step-up doses (0.06 and 0.3 mg/kg) on Days 2 and 4 followed by treatment doses (1.5 mg/kg) on Days 8 and 15 C2+: Q4W treatment doses (3 mg/kg) on Day 1 Daratumumab SC 1800 mg C1-2: weekly on Days 1, 8, 15, and 22 C3-6: Q2W on Days 1 and 15 C7+: Q4W on Day 1 Lenalidomide oral 25 mg* C2+: D1-21 Dexamethasone oral/IV 20 mg** C2-3: weekly on Days 1, 8, 15, and 22 * Lenalidomide dose may need to be adjusted for participants with reduced CrCl. ** Dexamethasone (16 mg) will be administered as pretreatment medication on Days 1, 2, 4, and 8 of Cycle 1. [0244] According to certain embodiments, a subject’s risk of high-grade CRS is reduced when the regimen comprises the initiation of the BCMAxCD3 bispecific antibody administration in Cycle 1 followed by the initiation of the lenalidomide administration in Cycle 2, compared to a regimen that comprises initiation of the lenalidomide administration in Cycle 1 followed by initiation of the BCMAxCD3 bispecific antibody administration in Cycle 2. [0245] According to certain embodiments, a subject’s risk of high-grade CRS is reduced when the regimen comprises the initiation of the BCMAxCD3 bispecific antibody administration in Cycle 1 followed by the initiation of the lenalidomide administration and the dexamethasone administration in Cycle 2, compared to a regimen that comprises initiation of the lenalidomide administration and the dexamethasone administration in Cycle 1 followed by initiation of the BCMAxCD3 bispecific antibody administration in Cycle 2. [0246] In certain embodiments, the regimen is a combination therapy comprising administration of the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab and lenalidomide. In certain embodiments, administration of the daratumumab starts in Cycle 1, administration of the BCMAxCD3 bispecific antibody starts in Cycle 1, and administration of the lenalidomide starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 1, and wherein a treatment dose of the
Docket No.258199.062002 (JBI6858WOPCT1) BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 2. In certain embodiments, the regimen further comprises: in Cycles 2 and 3 only, orally or intravenously administering dexamethasone to the subject. [0247] In certain embodiments, the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 μg/kg (e.g., on Day 2), a second step-up dose of 300 μg/kg (e.g., on Day 4), and then weekly (QW) doses of 1500 μg/kg (e.g., on Days 8 and 15), and in Cycle 2 and all subsequent treatment cycles, subcutaneously administering a monthly (Q4W) dose of 3000 μg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 2 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle. In certain embodiments, the regimen further comprises: in Cycles 2-3, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). [0248] In certain embodiments, the method achieves a decrease in new infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles. [0249] In certain embodiments, the method achieves a decrease in new grade ≥3 infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles.
Docket No.258199.062002 (JBI6858WOPCT1) [0250] In certain embodiments, the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria. [0251] In certain embodiments, the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria. [0252] In certain embodiments, the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria. [0253] In certain embodiments, the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. [0254] In certain embodiments, the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. [0255] In certain embodiments, the method achieves a rate of sustained MRD-negative CR (≥12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy. [0256] In certain embodiments, the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. [0257] In certain embodiments, the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months. [0258] In certain embodiments, the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is
Docket No.258199.062002 (JBI6858WOPCT1) greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. [0259] In certain embodiments, the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years. [0260] In certain embodiments, the method achieves a rate of sustained MRD-negative CR (≥12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (≥12 months) is greater than a reference rate of sustained MRD-negative CR (≥12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. [0261] In certain embodiments, after about 50 months of treatment, the rate of sustained MRD-negative CR (≥12 months) is greater than the reference rate of sustained MRD- negative CR (≥12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%. [0262] In certain embodiments, said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi- weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle; and for the dexamethasone: in Cycle 1 and all subsequent treatment cycles, orally or intravenously administering 40 mg or 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22).
Docket No.258199.062002 (JBI6858WOPCT1) Exemplary Embodiments: Additional Methods of Treating Newly Diagnosed Multiple Myeloma [0263] According to certain embodiments, the subject is newly diagnosed with multiple myeloma and previously received ASCT. [0264] According to certain embodiments, the subject is newly diagnosed with multiple myeloma and has completed induction therapy followed by ASCT (with or without consolidation). [0265] According to certain embodiments, the subject is newly diagnosed with multiple myeloma and previously received 4 to 6 cycles of induction therapy followed by ASCT. [0266] According to certain embodiments, the subject is newly diagnosed with multiple myeloma and previously received 4 to 6 cycles of 3- or 4-drug induction therapy that includes a proteasome inhibitor and/or an IMiD with or without anti-CD38 monoclonal antibody and a single or tandem ASCT. [0267] According to certain embodiments, the subject has received post-ASCT consolidation for up to 2 cycles if the total number of induction plus consolidation cycles did not exceed 6. [0268] According to certain embodiments, the regimen is a combination therapy comprising administration of at least one other anti-multiple myeloma agent in addition to the BCMAxCD3 bispecific antibody. [0269] According to certain embodiments, the regimen is a combination therapy comprising administration of lenalidomide and the BCMAxCD3 bispecific antibody, wherein the BCMAxCD3 bispecific antibody is teclistamab. [0270] According to certain embodiments, the step-up phase is Cycle 1 and comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0271] According to certain embodiments, the step-up phase is Cycle 1 and comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule).
Docket No.258199.062002 (JBI6858WOPCT1) [0272] According to certain embodiments, the step-up phase is Cycle 1 and comprises subcutaneously administering to the subject step-up doses of the BCMAxCD3 bispecific antibody followed by two 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0273] According to certain embodiments, the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15 of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). [0274] According to certain embodiments, the regimen is a combination therapy comprising administration of lenalidomide and teclistamab. [0275] According to certain embodiments, the lenalidomide is administered in an amount of 10 mg once daily. [0276] According to certain embodiments, the lenalidomide is not administered in Cycle 1. [0277] According to certain embodiments, the lenalidomide is not administered in Cycle 1, and is administered in Cycle 2 and all subsequent cycles. [0278] According to certain embodiments, the lenalidomide is administered in an amount of 10 mg once daily and may be increased to 15 mg once daily on Cycle 5 Day 1 if 10 mg was tolerated by the subject. [0279] According to certain embodiments, the BCMAxCD3 bispecific antibody is administered for a finite duration of 26 cycles. [0280] According to certain embodiments, the BCMAxCD3 bispecific antibody is administered for a finite duration of 13 cycles. [0281] According to certain embodiments, the BCMAxCD3 bispecific antibody is administered in Cycles 14-26 only if the subject has achieved a complete response or stringent complete response (according to IMWG 2016 criteria) after 13 cycles. [0282] An exemplary embodiment of a dosing regimen comprising teclistamab, alone or in combination with lenalidomide, is provided in Table E below.
Docket No.258199.062002 (JBI6858WOPCT1) Table E: Exemplary Treatment Method Treatment Cycle Dose Schedule (28-day cycle) Teclistamab SC Cycle 1 Step-up Dose 1: 0.06 mg/kg on Day 1 (alone or in Step-up Dose 2: 0.3 mg/kg on Day 3 combination with Treatment Dose: 1.5 mg/kg on Days 8 and 15 Lenalidomide)* Cycle 2-26
a Treatment Dose: 3 mg/kg Q4W on Day 1 *Lenalidomide Cycle 2-4 10 mg daily for 28 days Cycle 5-26 15 mg daily for 28 days (if 10 mg was tolerated) a. Subjects that achieve a complete response or stringent complete response after 13 cycles of treatment discontinue teclistamab but continue lenalidomide for the second year. [0283] According to certain embodiments, the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria. [0284] According to certain embodiments, the method achieves a CR or sCR in the subject, as defined by IMWG (2016) response criteria. [0285] According to certain embodiments, the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria. [0286] According to certain embodiments, the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria in at least 40%, or at least 45%, at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75% of subjects in a population with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT). [0287] According to certain embodiments, the method achieves MRD negativity (10
-5) in at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85% of subjects in a population with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT). [0288] According to certain embodiments, the method comprises administration of teclistamab alone or administration of a combination therapy comprising teclistamab and lenalidomide, and wherein said method achieves greater progression-free survival (PFS) and/or greater 12-month MRD-negative complete response (CR) rate in a population of subjects with newly diagnosed multiple myeloma that have previously received autologous
Docket No.258199.062002 (JBI6858WOPCT1) stem cell transplant (ASCT) compared to a reference population of subjects with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT), said reference population having been administered a regimen comprising lenalidomide without a BCMAxCD3 bispecific antibody. [0289] According to certain embodiments, treatment regimens as described herein (e.g., regimens comprising Q4W administration of teclistamab starting from Cycle 2) are administered to a subject that has newly diagnosed multiple myeloma and is eligible for ASCT. According to certain embodiments, the subject has not previously received ASCT. [0290] According to certain embodiments, the subject is administered the BCMAxCD3 bispecific antibody (e.g., teclistamab) as part of an induction therapy. As used herein, an induction therapy refers to a subject’s first phase of treatment for multiple myeloma. [0291] According to certain embodiments, the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and one or more additional antimyeloma agents, as an induction therapy. [0292] According to certain embodiments, the subject is administered multiple cycles of the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, as an induction therapy. [0293] According to certain embodiments, the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and one or more additional antimyeloma agents selected from the group consisting of daratumumab, lenalidomide, dexamethasone, and any combination thereof, as an induction therapy. [0294] According to certain embodiments, the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, lenalidomide, and dexamethasone, as an induction therapy. [0295] According to certain embodiments, the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, as an induction therapy. [0296] According to certain embodiments, the subject is administered 6 cycles of the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, as an induction therapy. [0297] According to certain embodiments, the subject is administered 6 cycles of a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, lenalidomide, and dexamethasone, as an induction therapy.
Docket No.258199.062002 (JBI6858WOPCT1) [0298] According to certain embodiments, the subject is administered 6 cycles of a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, as an induction therapy. [0299] According to certain embodiments, the subject is administered the induction therapy prior to having an autologous stem cell transplant (ASCT). According to certain embodiments, the subject is administered the induction therapy and then the subject undergoes an autologous stem cell transplant (ASCT) [0300] According to certain embodiments, the subject is administered a maintenance therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, after receiving ASCT. According to certain embodiments, the subject is administered a maintenance therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab) and daratumumab after receiving ASCT. According to certain embodiments, the maintenance therapy comprises 6-20 treatment cycles, or 12-18 treatment cycles, or 12 treatment cycles. [0301] Additional Embodiments [0302] According to certain embodiments, teclistamab is administered, as part of a combination regimen with talquetamab, to subjects iwith RRMM who have previously received 1 to 4 prior lines of therapy including an anti-CD38 antibody (e.g., daratumumab) and lenalidomide. Such embodiments are described, for example, in WO2024/220682, which is incorporated by reference herein. According to certain embodiments, a subject is administered one or more step-up doses followed by one or more doses of 1.5 mg/kg of teclistamab in Cycle 1, and then 3 mg/kg Q4W treatment doses of teclistamab starting from Cycle 2. According to certain embodiments, a subject is administered teclistamab at step-up doses of 0.06 mg/kg and 0.3 mg/kg, and two doses of 1.5 mg/kg in Cycle 1, and then 3 mg/kg Q4W treatment dose starting from Cycle 2. According to certain embodiments, a subject is administered teclistamab at step-up doses of 0.06 mg/kg and 0.3 mg/kg, and two doses of 1.5 mg/kg in Cycle 1, and then 3 mg/kg Q4W treatment dose starting from Cycle 2; and talquetamab SC step-up doses of 0.01 mg/kg and 0.06 mg/kg, one dose of 0.4 mg/kg, and one dose of 0.8 mg/kg in Cycle 1, followed by 0.8 mg/kg SC Q2W treatment doses in Cycles 2-4, wherein, from Cycle 5, participants with response of confirmed VGPR or better can change to talquetamab Q4W; and from Cycle 7, participants with response of confirmed PR or better must be switched to talquetamab 0.8 mg/kg Q4W.
Docket No.258199.062002 (JBI6858WOPCT1) [0303] According to an embodiment, a method of treating multiple myeloma in a subject in need thereof comprises treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1)-(4) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1)-(4) are met if (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT, or (3) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide as induction therapy (e.g., before the subject receives ASCT), or (4) the subject has received 1 to 4 prior lines of therapy including daratumumab and lenalidomide, and the teclistamab is administered as part of a combination regimen that further comprises administration of talquetamab. [0304] According to an embodiment, a method of treating newly diagnosed multiple myeloma in a subject in need thereof comprises treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1)-(3) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1)-(3) are met if: (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT, or (3) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that
Docket No.258199.062002 (JBI6858WOPCT1) further comprises administration of daratumumab and lenalidomide as induction therapy (e.g., before the subject receives ASCT). [0305] According to an embodiment, a method of treating newly diagnosed multiple myeloma in a subject in need thereof comprises treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1) or (2) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1) or (2) are met if: (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT. Alternative Step-Up Dosing Regimens [0306] Embodiments of the present invention also provide alternative step-up dosing regimens for a BCMAxCD3 bispecific antibody such as teclistamab, which may confine CRS events to earlier time points, potentially shortening the CRS monitoring window. In the phase 1/2, single-arm MajesTEC-1 study, teclistamab demonstrated manageable safety with rapid, deep, and durable responses. In MajesTEC-1, patients received step-up doses (SUDs) of 0.06 and 0.3 mg/kg followed by 1.5 mg/kg weekly (recommended phase 2 dose). Cytokine release syndrome (CRS) occurred in 72% of patients (nearly all grade 1/2); although most events occurred during step up; 24% of patients experienced CRS after the first treatment dose. Higher SUDs were evaluated in exploratory phase 1 cohorts from MajesTEC-1: Cohort 22 (SUDs of 0.1 and 0.5 mg/kg with 2-4 days between doses) and Cohort 23 (SUDs of 0.2 and 0.7 mg/kg with 2-4 days between doses). Based on results described herein, it is believed that higher SUDs may confine CRS events to earlier time points, potentially shortening the CRS monitoring window, and facilitating prompt transition of care to the outpatient setting. [0307] According to an embodiment, a method of treating multiple myeloma in a subject in need thereof comprises administering to the subject a therapeutically effective amount of any of the BCMAxCD3 bispecific antibodies described herein (e.g., teclistamab), wherein the
Docket No.258199.062002 (JBI6858WOPCT1) method comprises subcutaneously administering only two step-up doses of the BCMAxCD3 bispecific antibody before subcutaneously administering a first treatment dose (e.g., a first treatment dose of 1.5 mg/kg), wherein the two step-up doses are administered either (i) in an amount of 0.1 mg/kg followed by an amount of 0.5 mg/kg, or (ii) in an amount of 0.2 mg/kg followed by an amount of 0.7 mg/kg. According to certain embodiments, the time between the two step-up doses is between about 2 days and about 4 days. After the step-up doses, the subject may be administered treatment doses of the the BCMAxCD3 bispecific antibody according to any of the treatment dosing regimens described herein. [0308] According to certain embodiments, the subject has relapsed or refractory multiple myeloma. According to certain embodiments, the subject has previously received at least one, at least two, at least three or at least four prior lines of therapy. According to certain embodiments, the subject has previously received at least three or at least four prior lines of therapy. According to certain embodiments, the subject has previously received 1-3 prior lines of therapy, e.g., including a minimum of two consecutive cycles of daratumumab and two consecutive cycles of lenalidomide. According to certain embodiments, the subject has previously received 1-3 prior lines of therapy, including a proteasome inhibitor (PI) and lenalidomide. According to certain embodiments, the subject has previously received 1 to 4 prior lines of therapy including daratumumab and lenalidomide. [0309] According to certain embodiments, the subject has newly diagnosed multiple myeloma. According to certain embodiments, the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy. According to certain embodiments, the subject has newly diagnosed multiple myeloma and previously received ASCT. According to certain embodiments, the subject has newly diagnosed multiple myeloma and has not yet received ASCT. Prophylactic administration of toci [0310] Embodiments of the present invention provide methods of treating multiple myeloma in a subject in need thereof by administering to the subject a therapeutically effective amount of tocilizumab prior to administering a first dose of the BCMAxCD3 bispecific antibody (e.g., teclistamab). [0311] In certain embodiments, a single dose of toci before teclistamab treatment reduces the incidence of CRS relative to the overall patient population with no new safety signals and no evidence of impact on response to teclistamab. In certain embodiments, prophylactic toci
Docket No.258199.062002 (JBI6858WOPCT1) reduces CRS risk in patients with disease profiles suitable for outpatient dosing, reducing the burden of hospitalization during teclistamab step-up dosing. [0312] In certain embodiments, toci is administered prior to teclistamab as a single 8 mg/kg IV dose. EXEMPLARY EMBODIMENTS [0313] Provided below are enumerated embodiments of the present invention. These embodiments are illustrative only and do not limit the scope of the present disclosure or of the claims attached hereto. 1. A method of treating multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein: one or more step-up doses of a BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase, and treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. 2. The method of embodiment 1, wherein the BCMAxCD3 bispecific antibody comprises a BCMA binding domain comprising the HCDR1 of SEQ ID NO: 4, the HCDR2 of SEQ ID NO: 5, the HCDR3 of SEQ ID NO: 6, the LCDR1 of SEQ ID NO: 7, the LCDR2 of SEQ ID NO: 8 and the LCDR3 of SEQ ID NO: 9, and a CD3 binding domain comprising the HCDR1 of SEQ ID NO: 14, the HCDR2 of SEQ ID NO: 15, the HCDR3 of SEQ ID NO: 16, the LCDR1 of SEQ ID NO: 17, the LCDR2 of SEQ ID NO: 18 and the LCDR3 of SEQ ID NO: 19. 3. The method of embodiment 1 or 2, wherein the BCMA binding domain comprises a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 11, and the CD3 binding domain comprises a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 20 and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO: 21. 4. The method of any of embodiments 1-3, wherein the BCMAxCD3 bispecific antibody is an IgG1, an IgG2, an IgG3 or an IgG4 isotype. 5. The method of any of embodiments 1-4, wherein the BCMAxCD3 bispecific antibody is an IgG4 isotype.
Docket No.258199.062002 (JBI6858WOPCT1) 6. The method of any of embodiments 1-5, wherein the BCMAxCD3 bispecific antibody comprises one or more substitutions in its Fc region. 7. The method of any of embodiments 1-6, wherein the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises Proline/Alanine/Alanine substitutions at amino acid positions 228/234/235, respectively, in its Fc region (according to EU index numbering). 8. The method of any of embodiments 1-7, wherein the BCMAxCD3 bispecific antibody is an IgG4 isotype and comprises F405L and R409K substitutions in its Fc region (according to EU index numbering). 9. The method of any of embodiments 1-8, wherein the Fc region of the BCMA binding arm comprises Proline/Alanine/Alanine substitutions at amino acid positions 228/234/235, respectively (according to EU index numbering). 10. The method of any of embodiments 1-9, wherein the Fc region of the CD3 binding arm comprises Proline/Alanine/Alanine substitutions at amino acid positions 228/234/235, respectively, in addition to F405L and R409K substitutions (according to EU index numbering). 11. The method of any of embodiments 1-10, wherein the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having the amino acid sequence of SEQ ID NO: 23. 12. The method of any of embodiments 1-10, wherein the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 90% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 90% identity to the amino acid sequence of SEQ ID NO: 23. 13. The method of any of embodiments 1-10, wherein the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 95% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 95% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 95% identity to the amino acid sequence of SEQ ID NO: 23. 14. The method of any of embodiments 1-10, wherein the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 98% identity to the amino acid
Docket No.258199.062002 (JBI6858WOPCT1) sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 98% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 98% identity to the amino acid sequence of SEQ ID NO: 23. 15. The method of any of embodiments 1-10, wherein the BCMAxCD3 bispecific antibody comprises a first heavy chain (HC1) having at least 99% identity to the amino acid sequence of SEQ ID NO: 12, a first light chain (LC1) having at least 99% identity to the amino acid sequence of SEQ ID NO: 13, a second heavy chain (HC2) having at least 99% identity to the amino acid sequence of SEQ ID NO: 22 and a second light chain (LC2) having at least 99% identity to the amino acid sequence of SEQ ID NO: 23. 16. The method of any of embodiments 1-15, wherein the BCMAxCD3 bispecific antibody is teclistamab. 17. The method of any of embodiments 1-16, wherein each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg or 3 mg/kg. 18. The method of any of embodiments 1-16, wherein each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 1.5 mg/kg. 19. The method of any of embodiments 1-16, wherein each treatment dose of the BCMAxCD3 bispecific antibody that is administered to the subject on the monthly dosing schedule (Q4W) is administered in an amount of 3 mg/kg. 20. The method of any of embodiments 1-19, wherein each treatment dose of the BCMAxCD3 bispecific antibody administered to the subject on the monthly dosing schedule (Q4W) is administered on Day 1 of each BCMAxCD3 treatment cycle. 21. The method of any of embodiments 1-20, wherein the regimen does not include any BCMAxCD3 treatment cycles in which the BCMAxCD3 bispecific antibody is administered to the subject on a bi-weekly dosing schedule (Q2W). 22. The method of any of embodiments 1-21, wherein the step-up phase is 28 days. 23. The method of any of embodiments 1-22, wherein 1-3 step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase. 24. The method of any of embodiments 1-23, wherein two step-up doses of the BCMAxCD3 bispecific antibody are administered to the subject during the step-up phase.
Docket No.258199.062002 (JBI6858WOPCT1) 25. The method of any of embodiments 1-24, wherein the step-up phase comprises a first step-up dose of 0.06 mg/kg, or 0.1 mg/kg, or 0.2 mg/kg of the BCMAxCD3 bispecific antibody. 26. The method of any of embodiments 1-25, wherein the step-up phase comprises a first step-up dose of 0.06 mg/kg, or 0.1 mg/kg, or 0.2 mg/kg of the BCMAxCD3 bispecific antibody and a second step-up dose of 0.3 mg/kg, or 0.5 mg/kg, or 0.7 mg/kg of the BCMAxCD3 bispecific antibody. 27. The method of any of embodiments 1-26, wherein one or more treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. 28. The method of any of embodiments 1-27, wherein two treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. 29. The method of any of embodiments 1-28, wherein three treatment doses of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during the step-up phase, in addition to the one or more step-up doses. 30. The method of any of embodiments 1-29, wherein the step-up phase comprises subcutaneously administering two step-up doses and two treatment doses of the BCMAxCD3 bispecific antibody. 31. The method of any of embodiments 1-29, wherein the step-up phase comprises subcutaneously administering two step-up doses and three treatment doses of the BCMAxCD3 bispecific antibody. 32. The method of any of embodiments 27-31, wherein the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 0.72 mg/kg or 1.5 mg/kg or 3.0 mg/kg. 33. The method of any of embodiments 27-31, wherein each of the one or more treatment doses of the BCMAxCD3 bispecific antibody administered during the step-up phase are administered in an amount of 1.5 mg/kg. 34. The method of any of embodiments 27-33, wherein two or three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). 35. The method of any of embodiments 27-33, wherein two treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW).
Docket No.258199.062002 (JBI6858WOPCT1) 36. The method of any of embodiments 27-33, wherein three treatment doses of the BCMAxCD3 bispecific antibody are administered during the step-up phase on a weekly dosing schedule (QW). 37. The method of any of embodiments 1-29, wherein the step-up phase comprises subcutaneously administering to the subject (i) a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg and a second treatment dose of 1.5 mg/kg; or (ii) a first step-up dose of 0.1 mg/kg, a second step-up dose of 0.5 mg/kg, a first treatment dose of 1.5 mg/kg and a second treatment dose of 1.5 mg/kg; or (iii) a first step-up dose of 0.2 mg/kg, a second step-up dose of 0.7 mg/kg, a first treatment dose of 1.5 mg/kg and a second treatment dose of 1.5 mg/kg;. 38. The method of embodiment 37, wherein the second step-up dose is administered 2-4 days after first step-up dose. 39. The method of embodiment 37, wherein the second step-up dose is administered 2 days after first step-up dose. 40. The method of any of embodiments 37-39, wherein the first treatment dose is administered 4-7 days after the second step-up dose. 41. The method of any of embodiments 37-39, wherein the first treatment dose is administered 5 days after the second step-up dose. 42. The method of any of embodiments 37-41, wherein the second treatment dose is administered 5-9 days after the first treatment dose. 43. The method of any of embodiments 37-41, wherein the second treatment dose is administered 7 days after the first treatment dose. 44. The method of any of embodiments 1-43, wherein the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15. 45. The method of any of embodiments 37-44, wherein the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg, a second step-up dose of 0.3 mg/kg, a first treatment dose of 1.5 mg/kg, a second treatment dose of 1.5 mg/kg and a third treatment dose of 1.5 mg/kg. 46. The method of embodiment 45, wherein the third treatment dose is administered 5-9 days after the second treatment dose. 47. The method of embodiment 45, wherein the third treatment dose is administered 7 days after the second treatment dose.
Docket No.258199.062002 (JBI6858WOPCT1) 48. The method of any of embodiments 1-47, wherein the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22. 49. The method of any of embodiments 1-48, wherein: the step-up phase comprises subcutaneously administering to the subject (i) a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15, or (ii) a first step-up dose of 0.1 mg/kg on Day 1, a second step-up dose of 0.5 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15, or (iii) a first step-up dose of 0.2 mg/kg on Day 1, a second step-up dose of 0.7 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 50. The method of any of embodiments 1-48, wherein: the step-up phase comprises subcutaneously administering to the subject a first step- up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22, or (ii) a first step-up dose of 0.1 mg/kg on Day 1, a second step-up dose of 0.5 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22, or (iii) a first step-up dose of 0.2 mg/kg on Day 1, a second step-up dose of 0.7 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8, a second treatment dose of 1.5 mg/kg on Day 15 and a third treatment dose of 1.5 mg/kg on Day 22; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 51. The method of any of embodiments 1-50, wherein the treatment doses of the BCMAxCD3 bispecific antibody are administered to the subject on the monthly dosing schedule (Q4W) regardless of whether, or to what extent, the subject exhibits a clinical response to the regimen.
Docket No.258199.062002 (JBI6858WOPCT1) 52. The method of any of embodiments 1-51, wherein the step-up phase is in Cycle 1 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 2 of the regimen. 53. The method of any of embodiments 1-52, wherein the regimen is a combination therapy comprising administration of other anti-multiple myeloma agents in addition to the BCMAxCD3 bispecific antibody. 54. The method of any of embodiments 1-53, wherein the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen, or later. 55. The method of any of embodiments 1-53, wherein the regimen is a combination therapy, the step-up phase is in Cycle 2 of the regimen, and the first BCMAxCD3 treatment cycle is in Cycle 3 of the regimen. 56. The method of any of embodiments 1-16, wherein: during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg and a fourth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. 57. The method of embodiment 56, wherein: the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose and the fourth dose are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter. 58. The method of any of embodiments 1-16, wherein: during the step-up phase, a first dose of 0.06 mg/kg, a second dose of 0.3 mg/kg, a third dose of 1.5 mg/kg, a fourth dose of 1.5 mg/kg and a fifth dose of 1.5 mg/kg of the BCMAxCD3 bispecific antibody (e.g., teclistamab) are subcutaneously administered to the subject, and a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject every 4 weeks (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase. 59. The method of embodiment 58, wherein:
Docket No.258199.062002 (JBI6858WOPCT1) the step-up phase is 28 days and occurs in Cycle 1 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 2 and on Day 1 of each subsequent treatment cycle thereafter. 60. The method of embodiment 56, wherein: the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose and the fourth dose of the BCMAxCD3 bispecific antibody are administered on Day 1, Day 3, Day 8 and Day 15 of Cycle 2, respectively, and the treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. 61. The method of embodiment 58, wherein: the regimen is a combination therapy, the step-up phase is 28 days and occurs in Cycle 2 of the regimen; the first dose, the second dose, the third dose, the fourth dose and the fifth dose are administered on Day 1, Day 3, Day 8, Day 15 and Day 22 of Cycle 1, respectively, and the treatment dose of 3 mg/kg is administered on Day 1 of Cycle 3 and on Day 1 of each subsequent treatment cycle thereafter. 62. The method of any of embodiments 1-53, wherein the regimen is a combination therapy comprising administration of the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab and lenalidomide. 63. The method of embodiment 62, wherein administration of the daratumumab starts in Cycle 1, administration of the lenalidomide starts in Cycle 1, and administration of the BCMAxCD3 bispecific antibody starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 2, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 3. 64. The method of embodiment 63, wherein the regimen further comprises: in Cycles 1 and 2 only, orally or intravenously administering dexamethasone to the subject. 65. The method of any of embodiments 62-64, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab):
Docket No.258199.062002 (JBI6858WOPCT1) in Cycle 2, subcutaneously administering a first step-up dose of 60 μg/kg (e.g., on Day 1 or Day 2), a second step-up dose of 300 μg/kg (e.g., on Day 3 or Day 4), and then weekly (QW) doses of 1500 μg/kg (e.g., on Days 8 and 15), and in Cycle 3 and all subsequent treatment cycles, subcutaneously administering a monthly (Q4W) dose of 3000 μg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle. 66. The method of embodiment 65, wherein the regimen further comprises: in Cycles 1-2, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). 67. The method of any of embodiments 62-66, wherein the method achieves a decrease in new infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles. 68. The method of any of embodiments 62-67, wherein the method achieves a decrease in new grade ≥3 infections over time in a population of subjects with newly diagnosed multiple myeloma, compared to a population of subjects with newly diagnosed multiple myeloma that are treated with the combination therapy but receive more frequent doses of the BCMAxCD3 bispecific antibody (e.g., QW and/or Q2W) in Cycle 3 and all subsequent treatment cycles. 69. The method of any of embodiments 62-68, wherein the subject has newly diagnosed multiple myeloma and is either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy.
Docket No.258199.062002 (JBI6858WOPCT1) 70. The method of any of embodiments 62-69, wherein the subject has newly diagnosed multiple myeloma and is ineligible for autologous stem cell transplant (ASCT) as initial therapy. 71. The method of any of embodiments 62-70, wherein the subject has newly diagnosed multiple myeloma and is not intended for autologous stem cell transplant (ASCT) as initial therapy. 72. The method of any of embodiments 62-71, wherein the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria. 73. The method of any of embodiments 62-72, wherein the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria. 74. The method of any of embodiments 62-73, wherein the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria. 75. The method of any of embodiments 62-74, wherein the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. 76. The method of any of embodiments 62-75, wherein the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. 77. The method of any of embodiments 62-76, wherein the method achieves a rate of sustained MRD-negative CR (≥12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy. 78. The method of any of embodiments 62-77, wherein the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either
Docket No.258199.062002 (JBI6858WOPCT1) ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. 79. The method of embodiment 78, wherein the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months. 80. The method of any of embodiments 62-79, wherein the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. 81. The method of embodiment 80, wherein the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years. 82. The method of any of embodiments 62-81, wherein the method achieves a rate of sustained MRD-negative CR (≥12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (≥12 months) is greater than a reference rate of sustained MRD-negative CR (≥12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. 83. The method of embodiment 82, wherein, after about 50 months of treatment, the rate of sustained MRD-negative CR (≥12 months) is greater than the reference rate of sustained MRD-negative CR (≥12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%.
Docket No.258199.062002 (JBI6858WOPCT1) 84. The method of any of embodiments 78-83, wherein said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle; and for the dexamethasone: in Cycle 1 and all subsequent treatment cycles, orally or intravenously administering 40 mg or 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). 85. The method of any of embodiments 1-53, wherein the regimen is a combination therapy comprising administration of the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab and lenalidomide. 86. The method of embodiment 85, wherein administration of the daratumumab starts in Cycle 1, administration of the BCMAxCD3 bispecific antibody starts in Cycle 1, and administration of the lenalidomide starts in Cycle 2; and wherein one or more step-up doses and at least one treatment dose of the BCMAxCD3 bispecific antibody are subcutaneously administered to the subject during a step-up phase in Cycle 1, and wherein a treatment dose of the BCMAxCD3 bispecific antibody is subcutaneously administered to the subject monthly (Q4W) starting in Cycle 2. 87. The method of embodiment 86, wherein the regimen further comprises: in Cycles 2 and 3 only, orally or intravenously administering dexamethasone to the subject. 88. The method of any of embodiments 85-87, wherein the regimen comprises: for the BCMA x CD3 bispecific antibody (e.g., teclistamab): in Cycle 1, subcutaneously administering a first step-up dose of 60 μg/kg (e.g., on Day 2), a second step-up dose of 300 μg/kg (e.g., on Day 4), and then weekly (QW) doses of 1500 μg/kg (e.g., on Days 8 and 15), and
Docket No.258199.062002 (JBI6858WOPCT1) in Cycle 2 and all subsequent treatment cycles, subcutaneously administering a monthly (Q4W) dose of 3000 μg/kg (e.g., on Day 1); for the daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 2 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle. 89. The method of embodiment 88, wherein the regimen further comprises: in Cycles 2-3, orally or intravenously administering 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). 90. The method of any of embodiments 85-89, wherein the subject has newly diagnosed multiple myeloma and is either ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. 91. The method of any of embodiments 85-89, wherein the subject has newly diagnosed multiple myeloma and is ineligible for autologous stem cell transplant (ASCT) as initial therapy. 92. The method of any of embodiments 85-89, wherein the subject has newly diagnosed multiple myeloma and is not intended for autologous stem cell transplant (ASCT) as initial therapy. 93. The method of any of embodiments 85-92, wherein the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria. 94. The method of any of embodiments 85-92, wherein the method achieves a CR, VGPR, or PR in the subject, as defined by IMWG (2016) response criteria. 95. The method of any of embodiments 85-92, wherein the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria. 96. The method of any of embodiments 85-95, wherein the method achieves a median progression-free survival (mPFS) of at least 80 months, or at least 90 months, or at least 100 months, or at least 110 months in a population of subjects with newly diagnosed
Docket No.258199.062002 (JBI6858WOPCT1) multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. 97. The method of any of embodiments 85-96, wherein the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria, in at least 60%, or at least 65%, or at least 70% of subjects in a population with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy. 98. The method of any of embodiments 85-97, wherein the method achieves a rate of sustained MRD-negative CR (≥12 months) of at least about 18%, or at least about 20%, or at least about 22%, or at least about 24%, or at least about 26% in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy after about 50 months of treatment with the combination therapy. 99. The method of any of embodiments 85-98, wherein the method achieves a median progression-free survival (mPFS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mPFS is greater than a reference mPFS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. 100. The method of embodiment 99, wherein the mPFS is greater than the reference mPFS by at least about 10 months, or at least 20 months, or at least 30 months, or at least 40 months, or at least 50 months. 101. The method of any of embodiments 85-100, wherein the method achieves a median overall survival (mOS) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein the mOS is greater than a reference mOS achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide.
Docket No.258199.062002 (JBI6858WOPCT1) 102. The method of embodiment 101, wherein the mOS is greater than the reference mOS by at least 1 year, or at least 2 years, or at least 3 years, or at least 4 years. 103. The method of any of embodiments 85-102, wherein the method achieves a rate of sustained MRD-negative CR (≥12 months) in a population of subjects with newly diagnosed multiple myeloma that are ineligible or not intended for autologous stem cell transplant (ASCT) as initial therapy, wherein said rate of sustained MRD-negative CR (≥12 months) is greater than a reference rate of sustained MRD-negative CR (≥12 months) achieved in a reference population of subjects with newly diagnosed multiple myeloma who are either ineligible or not intended for ASCT as initial therapy, said reference population having been administered Daratumumab, Lenalidomide, and Dexamethasone (DRd) instead of the combination therapy comprising the BCMA x CD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide. 104. The method of embodiment 103, wherein, after about 50 months of treatment, the rate of sustained MRD-negative CR (≥12 months) is greater than the reference rate of sustained MRD-negative CR (≥12 months) by at least 4%, or at least 6%, or at least 8%, or at least 10%, or at least 12%, or at least 14%. 105. The method of any of embodiments 85-104, wherein said reference population has been administered the Daratumumab, Lenalidomide, and Dexamethasone (DRd) according to the following schedule: for the Daratumumab: in Cycles 1-2, subcutaneously administering 1800 mg weekly (QW) (e.g., on Days, 1, 8, 15 and 22), in Cycles 3-6, subcutaneously administering 1800 mg bi-weekly (Q2W) (e.g., on Days 1 and 15) and in Cycle 7 and all subsequent treatment cycles, subcutaneously administering 1800 mg monthly (Q4W) (e.g., on Day 1); and for the lenalidomide: in Cycle 1 and all subsequent treatment cycles, orally administering 25 mg daily for the first 21 days of each 28-day cycle; and for the dexamethasone: in Cycle 1 and all subsequent treatment cycles, orally or intravenously administering 40 mg or 20 mg of dexamethasone weekly (QW) (e.g., on Days 1, 8, 15 and 22). 106. The method of any of embodiments 85-105, wherein the subject’s risk of high- grade CRS is reduced when the regimen comprises the initiation of the BCMAxCD3
Docket No.258199.062002 (JBI6858WOPCT1) bispecific antibody administration in Cycle 1 followed by the initiation of the lenalidomide administration in Cycle 2, compared to a regimen that comprises initiation of the lenalidomide administration in Cycle 1 followed by initiation of the BCMAxCD3 bispecific antibody administration in Cycle 2. 107. The method of any of embodiments 85-105, wherein the subject’s risk of high- grade CRS is reduced when the regimen comprises the initiation of the BCMAxCD3 bispecific antibody administration in Cycle 1 followed by the initiation of the lenalidomide administration and the dexamethasone administration in Cycle 2, compared to a regimen that comprises initiation of the lenalidomide administration and the dexamethasone administration in Cycle 1 followed by initiation of the BCMAxCD3 bispecific antibody administration in Cycle 2. 108. The method of any of embodiments 1-50, wherein the subject has newly diagnosed multiple myeloma. 109. The method of any of embodiments 1-50, wherein the subject is newly diagnosed with multiple myeloma and previously received ASCT. 110. The method of any of embodiments 1-50, wherein the subject is newly diagnosed with multiple myeloma and has completed induction therapy followed by ASCT (with or without consolidation). 111. The method of any of embodiments 1-50, wherein the subject is newly diagnosed with multiple myeloma and previously received 4 to 6 cycles of induction therapy followed by ASCT. 112. The method of any of embodiments 1-50, wherein the subject is newly diagnosed with multiple myeloma and previously received 4 to 6 cycles of 3- or 4-drug induction therapy that includes a proteasome inhibitor and/or an IMiD with or without anti- CD38 monoclonal antibody and a single or tandem ASCT. 113. The method of any of embodiments 108-112, wherein the subject has received post-ASCT consolidation for up to 2 cycles if the total number of induction plus consolidation cycles did not exceed 6. 114. The method of any of embodiments 108-113, wherein the regimen is a combination therapy comprising administration of at least one other anti-multiple myeloma agent in addition to the BCMAxCD3 bispecific antibody. 115. The method of any of embodiments 108-114, wherein the regimen is a combination therapy comprising administration of lenalidomide and the BCMAxCD3 bispecific antibody.
Docket No.258199.062002 (JBI6858WOPCT1) 116. The method of embodiment 115, wherein the BCMAxCD3 bispecific antibody is teclistamab. 117. The method of any of embodiments 108-116, wherein the step-up phase occurs in Cycle 1 and comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 118. The method of any of embodiments 108-117, wherein the step-up phase occurs in Cycle 1 and comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 119. The method of any of embodiments 108-118, wherein the step-up phase occurs in Cycle 1 and comprises subcutaneously administering to the subject step-up doses of the BCMAxCD3 bispecific antibody followed by two 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 120. The method of any of embodiments 108-119, wherein the step-up phase comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15 of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 121. The method of any of embodiments 108-120, wherein the regimen is a combination therapy comprising administration of lenalidomide and the BCMAxCD3 bispecific antibody, and wherein the BCMAxCD3 bispecific antibody is teclistamab. 122. The method of embodiment 121, wherein the lenalidomide is administered in an amount of 10 mg once daily.
Docket No.258199.062002 (JBI6858WOPCT1) 123. The method of embodiment 121 or 122, wherein the lenalidomide is not administered in Cycle 1. 124. The method of any of embodiments 121-123, wherein the lenalidomide is not administered in Cycle 1, and is administered in Cycle 2 and all subsequent cycles. 125. The method of any of embodiments 121-124, wherein the lenalidomide is administered in an amount of 10 mg once daily and may be increased to 15 mg once daily on Cycle 5 Day 1 if 10 mg was tolerated by the subject. 126. The method of any of embodiments 121-125, wherein, the BCMAxCD3 bispecific antibody is administered for a finite duration of 26 cycles. 127. The method of any of embodiments 121-125, wherein, the BCMAxCD3 bispecific antibody is administered for a finite duration of 13 cycles. 128. The method of any of embodiments 121-125, wherein the BCMAxCD3 bispecific antibody is administered in Cycles 14-26 only if the subject has achieved a complete response or stringent complete response (according to IMWG 2016 criteria) after 13 cycles. 129. The method of any of embodiments 108-128, wherein the method achieves a clinical response in the subject that is a sCR, a CR, a VGPR, or a PR in the subject, as defined by IMWG (2016) response criteria. 130. The method of any of embodiments 108-128, wherein the method achieves a CR or sCR in the subject, as defined by IMWG (2016) response criteria. 131. The method of any of embodiments 108-128, wherein the method achieves a VGPR or PR in the subject, as defined by IMWG (2016) response criteria. 132. The method of any of embodiments 108-128, wherein the method achieves a CR or better (i.e., a sCR or a CR), as defined by IMWG (2016) response criteria in at least 40%, or at least 45%, at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75% of subjects in a population with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT). 133. The method of any of embodiments 108-128, wherein the method achieves MRD negativity (10
-5) in at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85% of subjects in a population with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT). 134. The method of any of embodiments 108-128, wherein the method comprises administration of teclistamab alone or administration of a combination therapy comprising
Docket No.258199.062002 (JBI6858WOPCT1) teclistamab and lenalidomide, and wherein said method achieves greater progression-free survival (PFS) and/or greater 12-month MRD-negative complete response (CR) rate in a population of subjects with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT) compared to a reference population of subjects with newly diagnosed multiple myeloma that have previously received autologous stem cell transplant (ASCT), said reference population having been administered a regimen comprising lenalidomide without a BCMAxCD3 bispecific antibody. 135. The method of any of embodiments 1-50, wherein the subject has newly diagnosed multiple myeloma and is eligible for ASCT. 136. The method of embodiment 135, wherein the subject has not previously received ASCT. 137. The method of embodiment 135 or 136, wherein the subject is administered the BCMAxCD3 bispecific antibody (e.g., teclistamab) as part of an induction therapy. 138. The method of any of embodiments 135-137, wherein the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and one or more additional antimyeloma agents, as an induction therapy. 139. The method of any of embodiments 135-138, wherein the subject is administered multiple cycles of the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, as an induction therapy. 140. The method of any of embodiments 135-139, wherein the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and one or more additional antimyeloma agents selected from the group consisting of daratumumab, lenalidomide, dexamethasone, and any combination thereof, as an induction therapy. 141. The method of any of embodiments 135-140, wherein the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, lenalidomide, and dexamethasone, as an induction therapy. 142. The method of any of embodiments 135-140, wherein the subject is administered a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, as an induction therapy. 143. The method of any of embodiments 135-142, wherein the subject is administered 4-8 cycles, preferably 6 cycles, of the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, as an induction therapy.
Docket No.258199.062002 (JBI6858WOPCT1) 144. The method of any of embodiments 135-143, wherein the subject is administered 6 cycles of a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, lenalidomide, and dexamethasone, as an induction therapy. 145. The method of any of embodiments 135-144, wherein the subject is administered 4-8 cycles, preferably 6 cycles, of a combination therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), daratumumab, and lenalidomide, as an induction therapy. 146. The method of any of embodiments 137-145, wherein the subject is administered the induction therapy prior to having an ASCT. 147. The method of any of embodiments 135-146, wherein the first cycle comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 148. The method of any of embodiments 135-146, wherein the first cycle comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 149. The method of any of embodiments 135-146, wherein the first cycle comprises subcutaneously administering to the subject step-up doses of the BCMAxCD3 bispecific antibody followed by two 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 150. The method of any of embodiments 135-146, wherein the first cycle comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15 of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a
Docket No.258199.062002 (JBI6858WOPCT1) treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 151. The method of embodiment 135, wherein the subject is administered a maintenance therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab), and optionally one or more additional antimyeloma agents, after receiving ASCT. 152. The method of embodiment 135, wherein the subject is administered a maintenance therapy comprising the BCMAxCD3 bispecific antibody (e.g., teclistamab) and daratumumab after receiving ASCT. 153. The method of embodiment 151 or 152, wherein the maintenance therapy comprises 6-20 treatment cycles. 154. The method of embodiment 151 or 152, wherein the maintenance therapy comprises 12-18 treatment cycles. 155. The method of embodiment 151 or 152, wherein the maintenance therapy comprises 12 treatment cycles. 156. The method of any of embodiments 151-155, wherein the first cycle comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 157. The method of any of embodiments 151-155, wherein the first cycle comprises subcutaneously administering to the subject one or more step-up doses of the BCMAxCD3 bispecific antibody followed by one or more 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 158. The method of any of embodiments 151-155, wherein the first cycle comprises subcutaneously administering to the subject step-up doses of the BCMAxCD3 bispecific antibody followed by two 1.5 mg/kg doses of the BCMAxCD3 bispecific antibody on a QW dosing schedule; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 159. The method of any of embodiments 151-155, wherein the first cycle comprises subcutaneously administering to the subject a first step-up dose of 0.06 mg/kg on Day 1, a
Docket No.258199.062002 (JBI6858WOPCT1) second step-up dose of 0.3 mg/kg on Day 3, a first treatment dose of 1.5 mg/kg on Day 8 and a second treatment dose of 1.5 mg/kg on Day 15 of the BCMAxCD3 bispecific antibody; and each subsequent BCMAxCD3 treatment cycle comprises subcutaneously administering a treatment dose of 3 mg/kg of the BCMAxCD3 bispecific antibody on Day 1 (i.e., on the Q4W dosing schedule). 160. A method of treating multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1)-(4) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1)-(4) are met if (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT, or (3) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide as induction therapy (e.g., before the subject receives ASCT), or (4) the subject has received 1 to 4 prior lines of therapy including daratumumab and lenalidomide, and the teclistamab is administered as part of a combination regimen that further comprises administration of talquetamab. 161. A method of treating newly diagnosed multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1)-(3) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1)-(3) are met if: (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of
Docket No.258199.062002 (JBI6858WOPCT1) daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT, or (3) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide as induction therapy (e.g., before the subject receives ASCT). 162. A method of treating newly diagnosed multiple myeloma in a subject in need thereof, comprising treating the subject according to a therapeutically effective regimen that comprises sequential 28-day BCMAxCD3 treatment cycles, wherein, if criteria (1) or (2) are met, then one or more step-up doses of teclistamab are subcutaneously administered to the subject during a step-up phase (e.g., during Cycle 1) and treatment doses of the teclistamab are subcutaneously administered to the subject on a monthly dosing schedule (Q4W) starting from the first BCMAxCD3 treatment cycle after the step-up phase (e.g., starting from Cycle 2), wherein the criteria (1) or (2) are met if: (1) the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy, and the teclistamab is administered as part of a combination regimen that further comprises administration of daratumumab and lenalidomide, or (2) the subject has newly diagnosed multiple myeloma, and the teclistamab is administered as part of a combination regimen that further comprises administration of lenalidomide after the subject received induction therapy followed by ASCT. 163. The method of any of embodiments 1-162, wherein the step-up phase comprises administering only two step-up doses of the BCMAxCD3 bispecific antibody (e.g., teclistamab) before administering a first treatment dose, wherein the two step-up doses are administered either (i) in an amount of 0.1 mg/kg followed by an amount of 0.5 mg/kg, or (ii) in an amount of 0.2 mg/kg followed by an amount of 0.7 mg/kg. 164. A method of treating multiple myeloma in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a BCMAxCD3 bispecific antibody (e.g., teclistamab), wherein the method comprises subcutaneously administering only two step-up doses of the BCMAxCD3 bispecific antibody before subcutaneously administering a first treatment dose, wherein the two step-up doses are administered either (i) in an amount of 0.1 mg/kg followed by an amount of 0.5 mg/kg, or (ii) in an amount of 0.2 mg/kg followed by an amount of 0.7 mg/kg. 165. The method of embodiment 164, wherein the two step-up doses are administered in an amount of 0.1 mg/kg followed by an amount of 0.5 mg/kg.
Docket No.258199.062002 (JBI6858WOPCT1) 166. The method of embodiment 164, wherein the two step-up doses are administered in an amount of 0.2 mg/kg followed by an amount of 0.7 mg/kg. 167. The method of any of embodiments 163-166, wherein the time between the two step-up doses is between about 2 days and about 4 days. 168. The method of any of embodiments 164-166, wherein the subject has relapsed or refractory multiple myeloma. 169. The method of embodiment 168, wherein the subject has previously received at least one, at least two, at least three or at least four prior lines of therapy. 170. The method of embodiment 168, wherein the subject has previously received 1-3 prior lines of therapy, e.g., including a minimum of two consecutive cycles of daratumumab and two consecutive cycles of lenalidomide. 171. The method of embodiment 168, wherein the subject has previously received 1-3 prior lines of therapy, including a proteasome inhibitor (PI) and lenalidomide. 172. The method of embodiment 168, wherein the subject has previously received 1 to 4 prior lines of therapy including daratumumab and lenalidomide. 173. The method of embodiment 168, wherein the subject has newly diagnosed multiple myeloma. 174. The method of embodiment 168, wherein the subject has newly diagnosed multiple myeloma and is ineligible or not intended for ASCT as initial therapy. 175. The method of embodiment 168, wherein the subject has newly diagnosed multiple myeloma and previously received ASCT. 176. The method of embodiment 168, wherein the subject has newly diagnosed multiple myeloma and has not yet received ASCT. [0314] Those skilled in the art will appreciate that numerous changes and modifications can be made to the preferred embodiments of the invention and that such changes and modifications can be made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention. [0315] The disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated herein by reference, in its entirety.
Docket No.258199.062002 (JBI6858WOPCT1) EXAMPLES [0316] The following examples are provided to further describe some of the embodiments disclosed herein. The examples are intended to illustrate, not to limit, the disclosed embodiments. Example 1: A Phase 3 Randomized Study Comparing Teclistamab in Combination with Daratumumab SC and Lenalidomide (Tec-DR) versus Daratumumab SC, Lenalidomide, and Dexamethasone (DRd), and Talquetamab in Combination with Daratumumab SC and Lenalidomide (Tal-DR) versus DRd, in Patients with Newly Diagnoses Multiple Myeloma Who are Either Ineligible or not Intended for Autologous Stem Cell Transplant as Initial Therapy (MajesTEC-7) [0317] A Phase 3 randomized study comparing teclistamab in combination with daratumumab SC and lenalidomide (Tec-DR) versus daratumumab SC, lenalidomide, and dexamethasone (DRd), and Talquetamab in Combination with Daratumumab SC and Lenalidomide (Tal-DR) versus DRd, will be carried out (NCT05552222). The Tal-DR arm of the study was added after initiation of the safety run-in phase of the study. The overall aim is to compare the efficacy between Tec-DR and DRd, and Tal-DR and DRd, in terms of PFS (progression-free survival, which refers to duration from the date of randomization to either progressive disease or death, whichever comes first) and minimal residual disease (MRD)- negative complete response (CR). [0318] Antibodies [0319] Anti-BCMA/anti-CD3 antibody teclistamab (also called Tec) (e.g., described in WO2017031104A1, the content of which is incorporated herein by reference in its entirety) was made by Janssen Pharmaceuticals. Teclistamab comprises a BCMA binding arm BCMB69 and a CD3 binding arm CD3B219, the amino acid sequences of which are shown in Table 5 and Table 6, respectively. Table 5. Sequences of BCMA binding arm of Teclistamab Region Sequence SEQ ID NO: BCMB69 HCDR1 SGSYFWG 4 HCDR2 SIYYSGITYYNPSLKS 5 HCDR3 HDGAVAGLFDY 6 LCDR1 GGNNIGSKSVH 7
Docket No.258199.062002 (JBI6858WOPCT1) LCDR2 DDSDRPS 8 LCDR3 QVWDSSSDHVV 9 VH QLQLQESGPGLVKPSETLSLTCTVSGGSISSGSY 10 FWGWIRQPPGKGLEWIGSIYYSGITYYNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCAR HDGAVAGLFDYWGQGTLVTVSS VL SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVH 11 WYQQPPGQAPVVVVYDDSDRPSGIPERFSGSN SGNTATLTISRVEAGDEAVYYCQVWDSSSDHV VFGGGTKLTVL HC QLQLQESGPGLVKPSETLSLTCTVSGGSISSGSY 12 FWGWIRQPPGKGLEWIGSIYYSGITYYNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCAR HDGAVAGLFDYWGQGTLVTVSSASTKGPSVFP LAPCSRSTSESTAALGCLVKDYFPEPVTVSWNS GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL GTKTYTCNVDHKPSNTKVDKRVESKYGPPCPP CPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSQEDPEVQFNWYVDGVEVHNAKTKPRE EQFNSTYRVVSVLTVLHQDWLNGKEYKCKVS NKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEM TKNQVSLTCLVKGFYPSDIAVEWESNGQPENN YKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNV FSCSVMHEALHNHYTQKSLSLSLGK LC SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVH 13 WYQQPPGQAPVVVVYDDSDRPSGIPERFSGSN SGNTATLTISRVEAGDEAVYYCQVWDSSSDHV VFGGGTKLTVLGQPKAAPSVTLFPPSSEELQAN KATLVCLISDFYPGAVTVAWKGDSSPVKAGVE TTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSC QVTHEGSTVEKTVAPTECS
Docket No.258199.062002 (JBI6858WOPCT1) Table 6. Sequences of CD3 binding arm of Teclistamab Region Sequence SEQ ID NO: CD3B219 HCDR1 TYAMN 14 HCDR2 RIRSKYNNYATYYAASVKG 15 HCDR3 HGNFGNSYVSWFAY 16 LCDR1 RSSTGAVTTSNYAN 17 LCDR2 GTNKRAP 18 LCDR3 ALWYSNLWV 19 VH EVQLVESGGGLVQPGGSLRLSCAASGFTFNTY 20 AMNWVRQAPGKGLEWVARIRSKYNNYATYY AASVKGRFTISRDDSKNSLYLQMNSLKTEDTA VYYCARHGNFGNSYVSWFAYWGQGTLVTVSS VL QTVVTQEPSLTVSPGGTVTLTCRSSTGAVTTSN 21 YANWVQQKPGQAPRGLIGGTNKRAPGTPARFS GSLLGGKAALTLSGVQPEDEAEYYCALWYSNL WVFGGGTKLTVLGQP HC EVQLVESGGGLVQPGGSLRLSCAASGFTFNTY 22 AMNWVRQAPGKGLEWVARIRSKYNNYATYY AASVKGRFTISRDDSKNSLYLQMNSLKTEDTA VYYCARHGNFGNSYVSWFAYWGQGTLVTVSS ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYF PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS SVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRV ESKYGPPCPPCPAPEAAGGPSVFLFPPKPKDTL MISRTPEVTCVVVDVSQEDPEVQFNWYVDGVE VHNAKTKPREEQFNSTYRVVSVLTVLHQDWL NGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQ VYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVE WESNGQPENNYKTTPPVLDSDGSFLLYSKLTV DKSRWQEGNVFSCSVMHEALHNHYTQKSLSLS LGK
Docket No.258199.062002 (JBI6858WOPCT1) LC QTVVTQEPSLTVSPGGTVTLTCRSSTGAVTTSN 23 YANWVQQKPGQAPRGLIGGTNKRAPGTPARFS GSLLGGKAALTLSGVQPEDEAEYYCALWYSNL WVFGGGTKLTVLGQPKAAPSVTLFPPSSEELQA NKATLVCLISDFYPGAVTVAWKADSSPVKAGV ETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYS CQVTHEGSTVEKTVAPTECS [0320] Objectives and Endpoints Table 7. Objectives and endpoints of the phase 3 study. Objectives Endpoints Primary x To compare the efficacy of Tec-DR versus Dual Primary Endpoints: DRd and Tal-DR versus DRd x PFS x Sustained MRD-negative CR (duration ≥ 12 months) Secondary x To further compare the efficacy of Tec-DR x VGPR or better versus DRd x CR or better x MRD-negative CR x PFS2 x OS x To assess the safety and tolerability of x Incidence and severity of AEs, teclistamab when administered in combination laboratory results, and other with DR safety parameters x To characterize PK of teclistamab x PK parameters using population PK approach x To assess the immunogenicity of teclistamab x Presence of ADAs to teclistamab
Docket No.258199.062002 (JBI6858WOPCT1) Objectives Endpoints x To assess participant’s symptoms, functioning, x Change from baseline in and HRQoL with Tec-DR versus DRd symptoms, functioning, and HRQoL x Time to sustained worsening in symptoms, functioning, and overall HRQoL Exploratory x To evaluate and explore pharmacodynamics biomarkers of antimyeloma and immune activity in participants treated with Tec-DR and DRd x To explore the relationship between PK, pharmacodynamics activity, AEs, and clinical response in participants treated with Tec-DR and DRd. x To explore the relationship between MRD negativity with duration and depth of clinical response in participants treated with Tec-DR and DRd. x To explore predictive biomarkers of response and resistance, including prognostic and disease markers at baseline, during treatment, and in relation to efficacy parameters x To identify patient subgroups that could respond differently to Tec-DR and DRd. x To evaluate MRU x To explore time to symptomatic progression in both treatment arms x To explore time to next treatment Abbreviations: AE=adverse event; CR=complete response; IMWG=International Myeloma Working Group; mBCAM=membrane BCMA; MRD=minimal residual disease; rHuPH20=recombinant human hyaluronidase PH20 enzyme; PR=partial response; sBCMA=soluble BCMA; sCR=stringent complete response; VGPR=very good partial response [0321] Study Design [0322] This is a randomized, open-label, multicenter, Phase 3 study in adult participants with newly diagnosed multiple myeloma according to the IMWG diagnostic criteria who are ineligible or not intended for ASCT as initial therapy. Inclusion Criteria and Exclusion Criteria are provided below.
Docket No.258199.062002 (JBI6858WOPCT1) [0323] Inclusion Criteria: x Have a diagnosis of multiple myeloma according to the International Myeloma Working Group (IMWG) diagnostic criteria x Be newly diagnosed and not considered a candidate for high-dose chemotherapy with autologous stem cell transplant (ASCT) due to: ineligible due to advanced age OR; ineligible due to the presence of comorbid condition(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT OR; deferral of high- dose chemotherapy with ASCT as initial treatment x Have an Eastern Cooperative Oncology Group (ECOG) performance status score of 0 to 2 x A female participant must agree not to be pregnant, breastfeeding, or planning to become pregnant while enrolled in this study or within 3 months after the last dose of study treatment x A male participant must agree not to plan to father a child while enrolled in this study or within 3 months after the last dose of study treatment [0324] Exclusion Criteria: x Received a cumulative dose of systemic corticosteroids equivalent to greater than or equal to (>=) 20 milligrams (mg) of dexamethasone within 14 days before randomization x Had plasmapheresis within 28 days of randomization x Had a stroke, transient ischemic attack, or seizure within 6 months prior to randomization x Known allergies, hypersensitivity, or intolerance to teclistamab excipients x Known contraindications to the use of daratumumab or lenalidomide per local prescribing information [0325] Initial Study Interventions Prior to Modification Based On Clinical Data [0326] As described in the section below entitled “Updated Study Design Following Safety Run-In Cohort 1,” the study interventions described here were eventually modified, based on clinical data following initiation of the safety run-in phase of the study: - The treatments are administered in 28-day cycles. For Tec-DR cohorts: varying doses of Teclistamab (Tec) are administered with a constant dose of daratumumab (dara), lenalidomide (len), and dexamethasone. For DRd
Docket No.258199.062002 (JBI6858WOPCT1) cohorts: constant doses of daratumumab (dara), lenalidomide (len), and dexamethasone are administered. - For teclistamab fixed dosing, subjects are stratified into two cohorts based on weight. Subjects ≤ 60 kg receive a teclistamab SC step up doses of 3 mg and 15 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 100 mg on days 8, 15, and 22 of treatment cycle 1. Teclistamab SC 100 mg is administered weekly for the second treatment cycle, followed by 200 mg Q2W starting at treatment cycle 3, and 200 mg Q4W starting at treatment cycle 7. Subjects > 60 kg receive a teclistamab SC step up doses of 4 mg and 25 mg on days 2 and 4 of treatment cycle 1, followed by SC treatment doses of 150 mg on days 8, 15, and 22 of treatment cycle 1. Tec SC 150 mg is administered weekly for the second treatment cycle, followed by 300 mg Q2W starting at treatment cycle 3, and 300 mg Q4W starting at treatment cycle 7. - Daratumumab is administered to all subjects by SC injection at a dose of 1800 mg with 30000 units hyaluronidase to decrease injection volume required. Daratumumab SC is administered weekly in treatment cycles 1 and 2, Q2W in treatment cycles 3-6, and Q4W thereafter. - Lenalidomide is administered orally at 25 mg every day for 21 days of a 28- day treatment cycle. Lenalidomide treatment started at treatment cycle 2. - For the Tec-DR cohort, dexamethasone is administered orally or intravenously at 20 mg weekly. For the Tec-DR cohort in Safety Run In Cohort 1, dexamethasone is administered during treatment cycles 2, 3, and 4. For the DRd cohort, dexamethasone is administered at 40 mg, or if the participant is greater than 75 years of age or has a BMI less than 18.5 dexamethasone is administered at a dose of 20 mg. For the DRd cohort, dexamethasone is administered weekly for all cycles. - When daratumumab and teclistamab are administered on the same day, daratumumab is administered first. Step-up dose 1 of teclistamab is administered at least 20 hours after daratumumab SC. The first treatment dose of teclistamab is administered 3 hours after daratumumab. Subsequent treatment doses of teclistamab are administered at least 15 minutes after daratumumab SC. - The treatment also includes required and optional pretreatment medications. The pretreatment medications include dexamethasone (glucocorticoid), IV or
Docket No.258199.062002 (JBI6858WOPCT1) oral. Dexamethasone is administered at 16 mg on days 1, 2, 4 and 8 of treatment cycle 1. Dexamthasone is administered approximately 1 to 3 hours prior to daratumumab SC or teclistamab SC on days on which daratumumab SC is not administered. Additional pretreatment medications include diphenhydramine (anti-histamine) or an equivalent and acetaminophen (antipyretic). Diphenhydramine is administered at 25 mg to 50 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC. Diphenhydramine is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC. Acetaminophen is administered at 650 mg to 1000 mg on all days daratumumab SC is administered, on days of all step-up doses and first treatment dose of teclistamab SC. Acetaminophen is administered IV or oral 1 to 3 hours prior to daratumumab SC or teclistamab SC. Optionally, montelukast (10 mg) may be administered at the discretion of the examiner prior to administration of daratumumab. On days when daratumumab SC and teclistamab SC are given together, pretreatment medications should be given prior to daratumumab SC. If there are greater than 4 hours between administration of study drugs, a repeat dose of dexamethasone 8 mg must be given prior to the bispecific antibody. A participant treated with Tec-DR who experiences Grade ≥2 CRS/sARR related to teclistamab or daratumumab SC must receive dexamethasone 16 mg, diphenhydramine 25 to 50 mg or equivalent, and acetaminophen 650 to 1000 mg as pretreatment medication for at least the subsequent dose of the study drug to which the event was related. These event-driven pretreatment medications are not required if already being administered as planned pretreatment described above. Dexamethasone doses scheduled as background treatment for Cycles 2-4 in participants treated with Tec-DR can be used as event-driven pretreatment medication. [0327] Summary of Initial Trial Design Prior to Modification Based on Clinical Data [0328] Eligible patients will be aged ≥18 years with NDMM and are either ineligible or not intended for ASCT as initial therapy, have measurable disease, and an ECOG performance status score 0–2. A safety run-in phase for Tec-DR will precede the randomized phase. Approximately 1000 patients will be randomized 1:1 to receive Tec-DR or DRd. Treatment will continue until progressive disease, death, intolerable toxicity, loss to follow-up,
Docket No.258199.062002 (JBI6858WOPCT1) withdrawal of consent, or end of study, whichever occurs first. The dual primary endpoints are PFS and minimal residual disease (MRD)-negative complete response (CR) sustained for at least 12 months. Secondary endpoints include rate of CR or better, overall survival, rate of MRD-negative CR, rate of very good partial response or better, PFS2, patient-reported outcomes, and adverse events (AEs). Response will be assessed using 2016 International Myeloma Working Group criteria. AEs will be graded by Common Terminology Criteria for AEs v5.0, except for cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, which will be graded by American Society for Transplantation and Cellular Therapy guidelines. Results from this trial will provide insights into a possible new treatment regimen (Tec-DR) for patients with NDMM, which may provide improved outcomes. [0329] Updated Study Design Following Safety Run-In Cohort 1 [0330] Based on evolving data in the teclistamab clinical development program, it was decided to investigate modified dosing schedules following initiation of the Tec-DR Safety Run-In Cohort 1. As such, a second Tec-DR Safety Run-In Cohort 2 (n=30) will be performed in the current study with a modified schedule of teclistamab that includes a lead-in cycle of DRd, administration of teclistamab starting in Cycle 2 and Q4W dosing starting in Cycle 3. Additionally, a Talquetamab-DR (Tal-DR) regimen will be evaluated in Safety Run-in Cohort 3 (n=30). Subsequently, about 1500 participants (500 per arm) will be randomized 1:1:1 to Tec-DR, Tal-DR. [0331] The study treatment that was administered for the Tec-DR Safety Run-In Cohort 1 (Tec-DR SRI 1) is provided in Table 8. Participants enrolled in Safety Run-in Cohort 1 were initially treated with fixed dose teclistamab in combination with DR. Following implementation of a protocol amendment, due to a safety measure, all participants treated with fixed dose teclistamab were transitioned to weight-based teclistamab. Table 8. Study Treatment Administered – Tec-DR – Safety Run-in Cohort 1 Study Cycle (each Dose Schedule Treatment cycle=28 days) Teclistamab SC Cycle 1 Step-up Dose 1 (Day 2): 0.06 mg/kg Step-up Dose 2 (Day 4): 0.3 mg/kg Treatment Dose on Days 8, 15, and 22: 1.5 mg/kg Cycle 2 Treatment dose weekly on Days 1, 8, 15, and 22:
Docket No.258199.062002 (JBI6858WOPCT1) Study Cycle (each Dose Schedule Treatment cycle=28 days) 1.5 mg/kg Cycle 3-6 Treatment dose Q2W on Days 1 and 15: 3 mg/kg Cycle 7+ Treatment dose Q4W on Day 1: 3 mg/kg Daratumumab Cycles 1 and 2 Weekly dose on Days 1, 8, 15, 22 SC (1800 mg) Cycles 3 to 6 Q2W dose on Days 1 and 15 Cycles 7+ Q4W dosing on Day 1 Lenalidomide Cycle 1 No dosing PO (25 mg) Cycles 2+ Days 1-21 from Cycle 2 onwards Dexamethasone Cycles 2 to 4 Weekly dose on Days 1, 8, 15, and 22 PO/IV (20 mg) IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous [0332] For the randomized part of this study, the treatment to be administered to subjects in the DRd arm is provided in Table 9. Table 9. Study Treatment Administered – DRd – Randomized Part Study Treatment Cycle (each Dose Schedule cycle=28 days) Daratumumab SC Cycles 1 and 2 Weekly dose on Days 1, 8, 15, 22 (1800 mg) Cycles 3 to 6 Q2W dose on Days 1 and 15 Cycles 7+ Q4W dosing on Day 1 Lenalidomide PO Cycles 1+ Days 1-21 (25 mg)* Dexamethasone Cycles 1+ Weekly dose on Days 1, 8, 15, and 22 PO/IV (40 mg)** IV=intravenous; PO=per os (oral); Q2W=every other week; SC=subcutaneous * Lenalidomide dose may need to be adjusted for participants with reduced CrCl ** For participants >75 years of age or with BMI <18.5 kg/m2, dexamethasone may be administered at a dose of 20 mg. [0333] As of 10 April 2023, 26 participants with newly diagnosed multiple myeloma were enrolled in Tec-DR Safety Run-in Cohort 1 of the current study and completed at least 2
Docket No.258199.062002 (JBI6858WOPCT1) cycles of treatment with Tec-DR. The median time from diagnosis to enrollment was 1 month (range, 0.16-4.76 months). The median age of participants at time of consent was 72.5 years (range, 66-84 years), and the majority (n=21, 81%) of participants were 70 years of age or older. The majority (n=22, 85%) of participants were transplant ineligible due to advanced age, with the remaining 15% transplant deferred due to a desire for treatment with a novel therapy. Preliminary efficacy results for the TEC-DR combo are shown in Table 10 below, in particular, the results for "VGPR or better” (VGPR, CR or sCR) and “CR or better” (CR or sCR) response rates at median follow-up (mFU) of 6.5 months. Table 10. MajesTEC-7 SRI1 N=26 mFU=6.5 months Overall Response Rate (ORR) (%) 24/26 (92.3%) CR+ 9/26 (34.6%) VGPR+ 21/26 (80.7%) [0334] With regard to the safety profile observed in Tec-DR SRI 1, using a median follow- up of 6.7 months, the rate of any grade infection was 92.3%, the rate of Grade 3+ infections was 38.5% and the rate of Grade 3 or 4 infections was 34.6%. By further optimizing the dosing regimen, it may be possible to decrease the rates of infection, thereby improving the safety profile. [0335] Updated Safety/Efficacy Results for Tec-DR SRI 1: [0336] As of Nov 27, 2023, 26 pts had received tec + DR (median, 11 cycles; range, 2–14) and 24 pts (92.3%) remained on tx. Median follow-up was 10.2 mo (range, 2–12). At baseline, median age was 72.5 yrs, 11.5% had an ECOG PS score of 2, and 15.4% had ≥1 soft-tissue plasmacytoma.4 pts (15.4%) deferred transplant. Treatment-emergent AEs (TEAEs) occurred in 100% of pts (grade [gr] 3/4, 22 pts [84.6%]). Infections occurred in 25 pts (96.2%; gr 3/4, 8 pts [30.8%]). CRS occurred in 16 pts (61.5%; all gr 1). ICANS occurred in 1 pt (gr 1). Gr 3/4 TEAEs occurring in ≥3 pts were neutropenia (13 [50%]), febrile neutropenia (5 [19.2%]), thrombocytopenia (4 [15.4%]), COVID-19 (3 [11.5%]), maculo- papular rash (3 [11.5%]), and hypertension (3 [11.5%]).1 pt discontinued tec + DR due to withdrawal of consent.2 discontinued len due to TEAEs (gr 3 maculo-papular rash and gr 4 neutropenia). There was 1 death due to a TEAE in cycle 3 (pneumonia influenza). Overall
Docket No.258199.062002 (JBI6858WOPCT1) response rate was 92.3% (complete response or better, 73.1%; very good partial response or better, 92.3%). These results from the first SRI of MajesTEC-7 demonstrate a manageable safety profile with early efficacy of tec + DR in NDMM. [0337] Tec-DR Safety Run-In 2 and Initial Randomized Tec-DR Arm: [0338] The study treatment to be administered for the Tec-DR Safety Run-In Cohort 2 (Tec-DR SRI 2), and the initial Randomized Tec-DR Arm, is provided in Table 11. The aim of the dosing regimen used in Tec-DR SRI 2 is to achieve robust efficacy while improving the safety profile seen in Tec-DR SRI 1, including reduced rates of infections. Table 11. Tec-DR – Safety Run-In Cohort 2 and Initial Randomized Part Study Treatment Dose Schedule Teclistamab SC C2: 2 step-up doses (0.06 and 0.3 mg/kg) on Days 1 and 3 followed by treatment doses (1.5 mg/kg) on Days 8 and 15 C3+: Q4W treatment doses (3 mg/kg) on Day 1 Daratumumab SC 1800 mg C1-2: weekly on Days 1, 8, 15, and 22 C3-6: Q2W on Days 1 and 15 C7+: Q4W on Day 1 Lenalidomide oral 25 mg* C1+: D1-21 Dexamethasone oral/IV 20 mg C1-2: weekly on Days 1, 8, 15, and 22 * Lenalidomide dose may need to be adjusted for participants with renal insufficiency as determined by CrCl. [0339] As shown in Table 11, teclistamab will be given after a DR lead-in cycle, and at a reduced frequency (Q4W) after the step-up phase is complete. The early reduction in frequency of teclistamab (Q4W dosing at Cycle 3) was implemented with the aim of reducing the rate of severe infections. The teclistamab SC dose schedule for Safety Run-in Cohort 2 of the study will include 2 step-up doses (0.06 and 0.3 mg/kg) on Days 1 and 3 of Cycle 2 followed by 1.5 mg/kg on Days 8 and 15 of Cycle 2 and 3 mg/kg Q4W starting from Cycle 3. [0340] The modified dosing regimen of teclistamab to be evaluated in Safety Run-in Cohort 2 is supported by emerging data from participants with relapsed/refractory multiple myeloma treated with teclistamab monotherapy (MajesTEC-1; Study 64407564MMY1001). In updated results from MajesTEC-1 as of the clinical cutoff date of 04 January 2023, 63 responders who received the RP2D dose (1.5 mg/kg QW) switched to Q2W treatment
Docket No.258199.062002 (JBI6858WOPCT1) with a median time to switch of 11.3 months (range: 1.0 to 24.7) and 9 of those participants had switched from 1.5 mg/kg Q2W to 1.5 mg/kg Q4W dosing. Forty-nine (77.8%) of the 63 participants did not have progressive disease, with a median follow-up of 12.6 months after schedule change. For those participants who switched to 1.5 mg/kg Q2W dosing, median DOR was not reached, and the 24-month event free rate was 68.7% (95% CI: 53.6 to 79.7). [0341] Additionally, in the MajesTEC-1 study, among those who stayed on treatment for at least 12 months, patients who switched to 1.5 mg/kg Q2W by 12 months experienced a lower incidence of Grade ≥3 treatment-emergent infections between 12 to 18 months of treatment than those who remained on 1.5 mg/kg QW dosing by 12 months (15.6% vs 33.3%). [0342] Using a population PK model based on teclistamab clinical data, it is estimated that the 3 mg/kg Q4W dosing regimen will have a lower exposure (concentration-time profile) than 1.5 mg/kg QW at steady state, which has been associated with durable responses. It is also estimated that the 3 mg/kg Q4W dosing regimen will achieve comparable PK parameters at steady state (C
trough, C
max, and AUC) to that of 1.5 mg/kg Q2W dosing after switch from 1.5 mg/kg QW. Additionally, it is estimated that the modified Q4W regimen will resemble the exposure (concentration-time profile) of the 0.72 mg/kg QW dose, which has shown robust efficacy and good safety in combination with daratumumab and lenalidomide in MajesTEC-2 (Study 64007957MMY1004). [0343] UPDATED Study Design Following Safety Run-In Cohorts 1 and 2: [0344] Safety Run-In 2 (SRI2) introduced a DRd lead-in dosing strategy (i.e., Drd dosing in Cycle 1 and introduction of Tec in Cycle 2 with step-up dosing occurring while lenalidomide is dosed). This approach was adopted to explore whether a DRd lead-in would reduce the burden of disease in newly diagnosed patients and further reduce the occurrence and severity of CRS. However, the data suggest that a lead-in cycle of DRd does not result in a reduction of the severity of CRS events. In contrast, there may be a potentially increased risk of higher- grade CRS when participants are exposed to both prior lenalidomide during Cycle 1 and concurrent lenalidomide during administration of the step-up dosing of Tec, supporting the reversion for the Randomized Part to delayed start of lenalidomide at Cycle 2 (i.e., supporting a switch from the dosing schedule of the Initial Randomized Part shown in Table 11 to the dosing schedule of the Updated Randomized Part shown in Table 12 below). In addition, Grade 5 infections were reported in 4 participants in SRI2, which can be explained at least in part by the high dose steroid included in the DRd lead-in.
Docket No.258199.062002 (JBI6858WOPCT1) [0345] The prior dexamethasone exposure before having started Tec in SRI2 may have contributed as well to the infection findings. While the DRd lead-in dosing led to higher grade CRS (SRI1) and an increase in fatal infections (SRI2), early preliminary efficacy in these cohorts support the lower exposure of Tec adopted in these cohorts. Therefore, the dosing schedule in the Randomized Part minimizes exposure to Tec and optimizes tolerability and safety for participants by completing step-up dosing in Cycle 1 and transitioning to monthly dosing in Cycle 2. Teclistamab and daratumumab will be initiated in Cycle 1 and lenalidomide added in Cycle 2 after the step-up phase. The less frequent (i.e., Q4W) dosing implemented in SRI2 to mitigate infection is retained. The teclistamab SC dose schedule for the Updated Randomized Part of the study will include 2 step-up doses (0.06 and 0.3 mg/kg) on Days 2 and 4 of Cycle 1 followed by 1.5 mg/kg on Days 8 and 15 of Cycle 1. Starting from Cycle 2, dosing will be 3 mg/kg Q4W on Day 1 of the cycle. Table 12 provides an overview of administration of the study drugs for Tec-DR for participants in the Updated Randomized Part. Table 12. Study Treatment Administered – UPDATED Tec-DR Randomized Part Study Treatment Dose Schedule Teclistamab SC C1: 2 step-up doses (0.06 and 0.3 mg/kg) on Days 2 and 4 followed by treatment doses (1.5 mg/kg) on Days 8 and 15 C2+: Q4W treatment doses (3 mg/kg) on Day 1 Daratumumab SC 1800 C1-2: weekly on Days 1, 8, 15, and 22 mg C3-6: Q2W on Days 1 and 15 C7+: Q4W on Day 1 Lenalidomide oral 25 C2+: D1-21 mg* Dexamethasone oral/IV C2-3: weekly on Days 1, 8, 15, and 22 20 mg** * Lenalidomide dose may need to be adjusted for participants with reduced CrCl. ** Dexamethasone (16 mg) will be administered as pretreatment medication on Days 1, 2, 4, and 8 of Cycle 1.
Docket No.258199.062002 (JBI6858WOPCT1) Example 2: Phase 3 Study of Teclistamab in Combination With Lenalidomide and Teclistamab Alone Versus Lenalidomide Alone in Participants With Newly Diagnosed Multiple Myeloma as Maintenance Therapy Following Autologous Stem Cell Transplantation (MajesTEC-4; NCT05243797) [0346] Background & Eligibility Criteria [0347] This is a multicenter, randomized, open-label, Phase 3 study in participants with newly diagnosed multiple myeloma to evaluate the benefits of teclistamab in combination with lenalidomide and teclistamab alone versus lenalidomide alone as maintenance therapy after autologous stem cell transplant. [0348] Inclusion Criteria include the following: x Must have a new diagnosis of multiple myeloma according to IMWG criteria and have received induction +/- consolidation. x Must have received only one line of therapy and achieved at least a partial response (≥PR) as per IMWG 2016 response criteria (Kumar 2016) without evidence of progression at the time of first treatment dose. x Must not be intolerant to the starting dose of lenalidomide. x Must not have received any maintenance therapy. x Have an ECOG performance status score of 0, 1, or 2 at screening and immediately prior to the start of administration of study treatment x Have clinical laboratory values within prespecified range. [0349] Exclusion Criteria include the following: x Received any prior BCMA-directed therapy. x Any previous therapy with an immune cell redirecting agent or gene modified adoptive cell therapy (eg, chimeric antigen receptor modified T cells, NK cells). x Discontinued treatment due to any AE related to lenalidomide as determined by the investigator. x Progressed on multiple myeloma therapy at any time prior to screening. x Received a cumulative dose of corticosteroids equivalent to ≥140 mg of prednisone within the 14 days prior to first treatment dose. x Received a live, attenuated vaccine within 4 weeks before first treatment dose. Non- live vaccines or non-replicating authorized for emergency use (eg. COVID-19) are allowed.
Docket No.258199.062002 (JBI6858WOPCT1) [0350] Len maintenance after ASCT has significant progression-free and overall survival benefits and is considered standard-of-care for transplant-eligible NDMM. However, patients (pts) eventually relapse, supporting the need for novel maintenance strategies to improve outcomes. Tec, a first-in-class B-cell maturation antigen × CD3 bispecific antibody, demonstrated deep and durable responses in multiple myeloma (MM), leading to its approval for triple-class exposed relapsed and refractory MM. MajesTEC-4/EMN30 is a multicenter, randomized, open-label, Phase 3 study evaluating Tec-Len, Tec, and Len maintenance therapy in NDMM after induction and ASCT, ± consolidation. [0351] Objectives and Endpoints [0352] The primary objective of this study is to compare the efficacy of teclistamab in combination with lenalidomide (Tec-Len) with that of lenalidomide monotherapy (Len), and the efficacy of teclistamab monotherapy (Tec) with that of lenalidomide monotherapy (Len) in the maintenance setting, as assessed by PFS and 12-month MRD-negative CR. Secondary objectives include further comparison of efficacy including OS (key secondary endpoint), CR or better, MRD-negative CR, sustained MRD-negative CR and MRD-negative conversion, PFS after next line of therapy (PFS2), time to next treatment (TTNT), and also safety, PK, and immunogenicity. [0353] Safety Run-In Cohorts [0354] Prior to the start of the randomized portion of the study, a first safety run-in (SRI1) was conducted. Participants in SRI1 receive Tec-Len for a duration of 2 years or until confirmed progressive disease, death, intolerable toxicity, or consent withdrawal, whichever occurs first. The dose schedule for SRI1 is provided in Table F below. Safety evaluation was performed after at least 20 participants received at least 2 cycles of treatment. Upon review of the safety data, the IDMC recommended to continue the study unmodified. However, based on emerging data from MajesTEC-1 suggesting a reduction in new onset of severe infections and durable responses with less frequent treatment dosing, it was decided to further optimize the teclistamab dosing schedule. Therefore, a second safety run-in (SRI2) was conducted with Q4W teclistamab dosing from Cycle 2. SRI2 includes 2 cohorts, one with Tec-Len (SRI2 Tec-Len) and one with Tec (SRI2 Tec). A safety evaluation was performed after at least 20 participants received at least 2 cycles of treatment and recommended to proceed to the randomized portion of the study. During the randomized portion of the study, participants will be randomized 1:1:1 to receive Tec-Len (Arm A), Len (Arm B), or Tec (Arm C). Participants will receive study treatment continuously for 2 years
Docket No.258199.062002 (JBI6858WOPCT1) or until confirmed progressive disease, death, intolerable toxicity, or consent withdrawal, whichever occurs first. Participants receiving Tec-Len in SRI1, Tec-Len in SRI2, and in Arm A who achieve a confirmed CR or better after 13 cycles of treatment will discontinue teclistamab but continue lenalidomide for the second year. [0355] Description of Study Treatment [0356] Study treatment is administered on 28-day cycles for SRI1, SRI2, Tec-Len (Arm A), Len (Arm B), and Tec (Arm C). Treatment continues for 2 years (ie, 26 cycles). Participants in SRI1 were treated with both weight-based and fixed dose teclistamab in combination with lenalidomide per the protocol amendment(s) approved during their enrollment and participation and have all switched to the weight-based dosing schedule as detailed below. Participants in SRI2 are treated with weight-based SC teclistamab in combination with lenalidomide or teclistamab alone. Similarly, teclistamab in the randomized portion of the study will be administered SC using a weight-based dose schedule as detailed in Table F below. Table F: Dose Schedules Treatment Cycle Dose Schedule (28-day cycle) Teclistamab SC (SRI1) Cycle 1 Step-up Dose 1: 0.06 mg/kg on Day 1 Step-up Dose 2: 0.3 mg/kg on Day 3 Treatment Dose: 1.5 mg/kg on Days 8, 15, and 22 Cycle 2 Treatment Dose: 1.5 mg/kg weekly on Days 1, 8, 15, and 22 Cycle 3-6 Treatment Dose: 3 mg/kg Q2W on Days 1 and 15 Cycle 7-26
a Treatment Dose: 3 mg/kg Q4W on Day 1 Teclistamab SC Cycle 1 Step-up Dose 1: 0.06 mg/kg on Day 1 (SRI2, Arm A, and Step-up Dose 2: 0.3 mg/kg on Day 3 Arm C) Treatment Dose: 1.5 mg/kg on Days 8 and 15 Cycle 2-26
a Treatment Dose: 3 mg/kg Q4W on Day 1 Lenalidomide PO Cycle 2-4 10 mg daily for 28 days (SRI1, SRI2 Tec-Len Cycle 5-26 15 mg daily for 28 days (if 10 mg was tolerated) and Arm A) Lenalidomide PO Cycle 1-3 10 mg daily for 28 days
Docket No.258199.062002 (JBI6858WOPCT1) (Arm B) Cycle 4-26 15 mg daily for 28 days (if 10 mg was tolerated) a. Participants in SRI1, SRI2 Tec-Len, or in Arm A who achieve a confirmed CR or better after 13 cycles of treatment will discontinue teclistamab but continue lenalidomide for the second year. [0357] Initial safety run-in (SRI) results are provided below. [0358] Methods for the Safety Run-In Cohorts: Eligible pts were aged ≥18 y, had NDMM (per International Myeloma Working Group [IMWG] criteria), received 4-6 cycles of 3- or 4- drug induction that included a proteasome inhibitor and/or an immunomodulatory drug ± an anti-CD38 antibody and a single or tandem ASCT ± consolidation, and achieved a partial response or better per IMWG 2016 response criteria. Three cohorts at different Tec dose frequencies were evaluated: Cohort 1 (Tec-Len) with Tec dosing at 1.5 mg/kg QW for 2 cycles (C), followed by 3 mg/kg Q2W in C3-6, and 3 mg/kg Q4W in C7+; Cohort 2 (Tec- Len) with Tec dosing at 1.5 mg/kg on Days 8 and 15 in C1, followed by 3 mg/kg Q4W in C2+; and Cohort 3 (Tec) dosing at 1.5 mg/kg on Days 8 and 15 in C1, followed by 3 mg/kg Q4W in C2+. All pts received the same inpatient Tec step-up schedule in C1 (0.06 mg/kg; 0.3 mg/kg). Len 10 mg QD in C2-4 (if tolerated, 15 mg thereafter) was given in Tec-Len cohorts. Treatment duration was 2 y for all pts; in Tec-Len cohorts, Tec was stopped after 13 cycles of treatment if complete response or better (≥CR) was achieved. Adverse events (AEs) were graded per Common Terminology Criteria for Adverse Events v5.0. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded per American Society for Transplantation and Cellular Therapy guidelines. Investigator-assessed response was based on IMWG 2016 criteria. [0359] Results from the Safety Run-In Cohorts: Across 3 cohorts, 94 pts were enrolled (Cohort 1, n=32; Cohort 2, n=32; Cohort 3, n=30). At a median follow-up of 14.4, 5.0, and 4.9 mo, 97% of pts in each cohort (n=31, 31, 29, respectively) remained on treatment. Baseline characteristics were generally balanced between cohorts with a median age of 58-59 y. Pts in Cohort 1, 2, and 3 had received a median of 15, 6, and 6 maintenance cycles, respectively. Neutropenia and infections were the most common Grade 3/4 treatment- emergent AEs (TEAEs). Compared with Cohort 1, the cumulative incidence of any grade and Grade 3/4 neutropenia at 4 mo showed a decreased trend in Cohorts 2 and 3 with less frequent Tec dosing (Cohort 1: any grade/Grade 3/4 incidence, 69%/66%; Cohort 2: 44%/41%; Cohort 3: 37%/28%). A similar trend was observed for all grade infections with less frequent Tec dosing (Cohort 1: 78%; Cohort 2: 63%; Cohort 3: 61%). Among 68/94
Docket No.258199.062002 (JBI6858WOPCT1) (72.3%) pts who had any grade hypogammaglobulinemia, 63/68 (92.6%) received ≥1 dose of IVIg. The overall CRS rate was 43.6%, with 6.4% Grade 2 and no high-grade events. The CRS rate following the first treatment dose of Tec (1.5 mg/kg) was low at 7.4%. No ICANS were reported. TEAEs led to treatment discontinuations in 2 pts (1 each in Cohorts 2 and 3) and death in 1 pt (Cohort 2; due to COVID-19 during C1, prior to the start of Len). In Cohort 1, all 28 pts with MRD assessments at 12 mo were in MRD negative CR. Among MRD positive pts at study entry, 10 (100%) converted to MRD negative CR during treatment. Among 16 pts who had <CR at study entry, 100% achieved ≥CR during treatment. At the time of clinical cutoff, no pts had progressed on treatment. [0360] Conclusions from Safety Run-In Cohorts: Overall, Tec-Len and Tec can be safely administered as maintenance therapy following ASCT in NDMM. Cohorts 2 and 3 showed a trend for improved early safety outcomes with less frequent Tec dosing compared with Cohort 1. Tec-Len demonstrated deepening of responses and 100% MRD negative CR rate at 12 mo in Cohort 1 evaluable pts at the time of the initial cutoff. These data informed the randomized part of MajesTEC-4/EMN30. The randomized portion is proceeding with less frequent Tec dosing (3.0 mg/kg Q4W starting from Cycle 2). Example 3: Higher Teclistamab Step-up Dosing in Patients With Relapsed or Refractory Multiple Myeloma (RRMM): Results From the MajesTEC-1 Trial [0361] In the phase 1/2, single-arm MajesTEC-1 study, teclistamab demonstrated manageable safety with rapid, deep, and durable responses. In MajesTEC-1, patients received step-up doses (SUDs) of 0.06 and 0.3 mg/kg followed by 1.5 mg/kg weekly (recommended phase 2 dose). Cytokine release syndrome (CRS) occurred in 72% of patients (nearly all grade 1/2); although most events occurred during step up; 24% of patients experienced CRS after the first treatment dose. Higher SUDs were evaluated in exploratory phase 1 cohorts from MajesTEC-1: Cohort 22 (SUDs of 0.1 and 0.5 mg/kg with 2-4 days between doses) and Cohort 23 (SUDs of 0.2 and 0.7 mg/kg with 2-4 days between doses). Following SUDs, patients received 1.5 mg/kg on Days 1, 8, and 15 of Cycle 1 followed by 3 mg/kg every 4 weeks in subsequent cycles. Premedication requirements and permitted supportive treatments for CRS were the same as for the RP2D cohort. Prophylactic tocilizumab was not permitted. CRS was graded per Lee criteria and converted to American Society for Transplantation and Cellular Therapy (ASTCT) criteria to allow comparison with the RP2D. All adverse events (AEs) were graded according to Common Terminology
Docket No.258199.062002 (JBI6858WOPCT1) Criteria for Adverse Events (CTCAE) Version 4.03. Serum samples were collected to assess teclistamab pharmacokinetics (PK). [0362] A total of 21 patients were enrolled (Cohort 22: n=10; Cohort 23: n=11). Patient demographics and baseline disease characteristics were generally consistent with those of the RP2D population. Median follow-up was 8.6 months (range, 3.4-10.6) for Cohort 22 and 3.2 months (range, 0.1-6.0) for Cohort 23. Overall, 22 CRS events were reported in 16 patients (8 patients in each cohort); all events were grade 1/2. Three (14.3%; 1 in Cohort 22 and 2 in Cohort 23) patients experienced recurrent CRS events. The overall incidence and severity of CRS events were comparable to results with the RP2D SUD (median follow-up: 14.1 months [range, 0.3-24.4]). CRS events occurred primarily during SUDs. Median onset of CRS through Cycle 1 Day 1 was 2.0 days in Cohort 22 and 2.5 days in Cohort 23; median event duration was 2 days in both cohorts. All CRS events resolved, and none led to treatment discontinuation. All 8 (100%) patients in Cohort 22 and 7 of 8 (87.5%) patients in Cohort 23 who experienced CRS received supportive treatment Overall, 5 of 8 (62.5%) patients in Cohort 22 and 4 of 8 (50.0%) patients in Cohort 23 who experienced CRS received tocilizumab. Of the 2 patients who experienced recurrent CRS during step up, only 1 (Cohort 22; grade 2) received tocilizumab; the other patient (Cohort 23; grade 1) did not receive supportive treatment. The 1 patient in Cohort 23 who experienced recurrent CRS after Cycle 1 Day 1 did not receive tocilizumab but received corticosteroids. No new safety signals were observed in the higher SUD cohorts, with safety profiles comparable to those of the RP2D. Based on limited preliminary data, median maximal serum teclistamab concentrations following SUD administration were slightly higher in the higher SUD cohorts compared with the RP2D SUDs. Trough concentrations of patients primed with higher SUDs were comparable to that observed with the RP2D after the treatment dose of 1.5 mg/kg. [0363] A key takeaway from these cohorts is that higher teclistamab SUDs resulted in earlier occurrence of CRS events during treatment initiation, shortening the time needed for CRS monitoring, and potentially facilitating the transition to outpatient dosing. Higher SUDs resulted in slightly higher teclistamab serum concentrations during step up relative to the RP2D but did not have an impact on teclistamab exposure following treatment doses. Rates of CRS in the higher SUD cohorts were similar to those with the RP2D; all events were grade 1/2, most occurred after the first SUD (including both grade 2 events in Cohort 23), and none led to treatment discontinuation. No new safety signals were observed compared with the RP2D SUDs.