[go: up one dir, main page]
More Web Proxy on the site http://driver.im/

CN118785922A - DLL3 targeted cancer treatment - Google Patents

DLL3 targeted cancer treatment Download PDF

Info

Publication number
CN118785922A
CN118785922A CN202380022555.3A CN202380022555A CN118785922A CN 118785922 A CN118785922 A CN 118785922A CN 202380022555 A CN202380022555 A CN 202380022555A CN 118785922 A CN118785922 A CN 118785922A
Authority
CN
China
Prior art keywords
agent
dose
dll
day
administered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202380022555.3A
Other languages
Chinese (zh)
Inventor
N·哈希米萨拉威
M·米诺卡
A·戈德里克
X·陈
M·基斯特勒
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority claimed from PCT/US2023/063010 external-priority patent/WO2023164474A1/en
Publication of CN118785922A publication Critical patent/CN118785922A/en
Pending legal-status Critical Current

Links

Landscapes

  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention disclosed herein provides a method for treating DLL3 positive cancer or SCLC comprising administering an anti-DLL 3 agent alone or in combination with an anti-PD-L1 antibody and/or a chemotherapeutic agent to a subject in need thereof. Stepped administration or prolonged IV infusion of the anti-DLL 3 agent is also disclosed.

Description

DLL3 targeted cancer treatment
Cross Reference to Related Applications
The present application claims the benefits of U.S. provisional application Ser. Nos. 63/313,119 and 63/429,311, filed on day 2, month 23, 2022 and day 12, 1, 2022, respectively. The contents of these applications are incorporated herein by reference in their entirety.
Submitting sequences in XML files
The context of the following submitted XML files is incorporated herein by reference in its entirety: 43,375 bytes of an XML file named "10009-WO01-SEC_ SequenceListing"; created at 2023, month 2 and 13.
Technical Field
The present application relates to dosages and administration of DLL3 targeted cancer therapies, including combination therapies using PD-L1 targeted agents and/or chemotherapeutic agents.
Background
Delta-like 3 (DLL 3) is a type 1 transmembrane protein and a non-classical Notch ligand. DLL3 is a promising target for the development of T cell therapies because it is highly expressed on the cell surface of neuroendocrine tumors and minimally localized in normal tissues, primarily cytoplasmic localization (Owen et al J Hematol Oncol [ journal of blood and oncology ],12:61 (2019)). Neuroendocrine tumors typically originate in neuroendocrine cells and may occur in organs such as the lung, appendix, small intestine, rectum and pancreas. Small Cell Lung Cancer (SCLC) is a neuroendocrine carcinoma in which DLL3 is differentially expressed. Using Immunohistochemistry (IHC), 85% of SCLC tumors stained positive for DLL3 (pattern consistent with membranous and cytoplasmic expression). In contrast, low levels of DLL3 protein expression were detected in normal brain, islets and pituitary with cytoplasmic staining patterns (samenders et al SCI TRANSL MED [ science transformation medicine ]7:302ra136 (2015)).
SCLC is an invasive form of lung cancer, has a poor prognosis and limited treatment options, and represents about 10% -15% of lung cancer. Survival rates have remained low for decades, with only 5% of SCLC patients surviving for five years, largely due to the lack of new therapies to combat this form of lung cancer. SCLC is characterized by neuroendocrine differentiation, high growth fraction, fast doubling time and early establishment of extensive metastatic lesions. About one third of patients exhibit localized disease. Most patients present with extensive disease. These phases affect the available treatment regimens, where localized diseases are treated with chemotherapy and radiation therapy, and widespread diseases are treated with chemotherapy alone.
SCLC patients respond very well to first-line chemotherapy (including etoposide and cisplatin) and radiotherapy, but relapse very quickly. Approved therapies for recurrent disease, but for patients lacking available treatment options, chemoresistance develops. The prognosis of recurrent refractory conditions is very poor, with rapid disease progression and short term median survival of less than six months once the patient receives the trilinear treatment. Patients with extended SCLC (ES-SCLC) develop resistance and die of the disease at a median time of 10 to 12 months after diagnosis
AMG 757 (also known as taratamab (tarlatamab)) is a bispecific T cell adapterMolecules targeting DLL3 on cancer cells and CD3 on T cells. It was developed for the treatment of DLL3 positive cancers, such as SCLC and neuroendocrine prostate cancer (NEPC), and is being evaluated in clinical trials.
Efforts have been made to develop therapies for the treatment of DLL3 positive cancers such as SCLC, but the overall survival rate is still poor. There is an unmet medical need for developing therapies for treating DLL3 positive cancers such as SCLC.
Disclosure of Invention
Based on the disclosure provided herein, one of ordinary skill in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following examples (E).
E1: a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, wherein the anti-DLL 3 agent is administered at a dose of from 10mg to 100mg twice every three weeks.
E2: a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, wherein the anti-DLL 3 agent is administered at a dose of from 20mg to 200mg once every three weeks.
E3: a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, wherein the anti-DLL 3 agent is administered according to the following regimen: a) Administering the anti-DLL 3 agent in a first period, wherein (i) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of from 1mg to 200mg over a period of 2 days to 7 days, and (ii) after the continuous intravenous infusion, the anti-DLL 3 agent is administered by bolus intravenous infusion on day 8, day 15, or both days 8 and 15, and b) the anti-DLL 3 agent is administered according to any one of the following i) to iii): i) Starting on day 29 and once every two weeks thereafter, administering one or more subsequent doses of the anti-DLL 3 agent at a dose from 10mg to 100 mg; ii) starting on day 22 and thereafter twice every three weeks, administering one or more subsequent doses of the anti-DLL 3 agent at a dose from 10mg to 100 mg; and iii) starting on day 22 and thereafter administering once every three weeks, one or more subsequent doses of the anti-DLL 3 agent are administered at a dose from 20mg to 200 mg.
E4: a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered according to any one of the following a) to c): a) Administering the anti-DLL 3 agent at a dose of from 10mg to 100mg once every two weeks; b) Administering the anti-DLL 3 agent twice every three weeks at a dose of from 10mg to 100 mg; and c) administering the anti-DLL 3 agent at a dose of from 20mg to 200mg once every three weeks.
E5: the method of any one of E1 to E4, wherein the anti-DLL 3 positive cancer is Small Cell Lung Cancer (SCLC).
E6: the method of any one of E1-E5, wherein the anti-DLL 3 positive cancer is recurrent/refractory (RR) SCLC or generalized disease (ED) SCLC.
E7: the method of any one of E1 to E6, wherein the anti-DLL 3 agent is a bispecific T cell engaging antigen binding polypeptide comprising two binding domains: the first domain binds to human DLL3 and the second domain binds to human CD 3.
E8: the method of E7 wherein the DLL3 binding domain binds to an epitope of human DLL3 contained within the amino acid sequence of SEQ ID No. 29.
E9: the method of E7 or E8, wherein the DLL3 binding domain comprises (a) a heavy chain variable region (VH) comprising: (i) VH complementarity determining region 1 (CDR-H1) comprising the amino acid sequence of SEQ ID No. 1; (ii) CDR-H2 comprising the amino acid sequence of SEQ ID NO. 2; and (iii) CDR-H3 comprising the amino acid sequence of SEQ ID NO. 3; and (b) a light chain variable region (VL) comprising: (i) VL complementarity determining region 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO. 4; (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 5; and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 6.
E10: the method of any one of E7-E9, wherein the DLL3 binding domain comprises: (1) A VH comprising the amino acid sequence of SEQ ID No. 7 and a VL comprising the amino acid sequence of SEQ ID No. 8, or (2) a VH comprising the amino acid sequence of SEQ ID No. 11 and a VL comprising the amino acid sequence of SEQ ID No. 12.
E11: the method of any one of E7 to E10, wherein the VH and VL of the DLL3 binding domain are connected by a linker to form a single chain Fv (scFv).
E12: the method of any one of E7 to E11 wherein the DLL3 binding domain comprises the amino acid sequence of SEQ ID No. 9 or SEQ ID No. 13.
E13: the method of any one of E7 to E12, wherein the CD3 binding domain comprises: (a) VH comprising: CDR-H1 comprising the amino acid sequence of SEQ ID NO. 18, CDR-H2 comprising the amino acid sequence of SEQ ID NO. 19, and CDR-H3 comprising the amino acid sequence of SEQ ID NO. 20; and VL, comprising: CDR-L1 comprising the amino acid sequence of SEQ ID NO. 15, CDR-L2 comprising the amino acid sequence of SEQ ID NO. 16, and CDR-L3 comprising the amino acid sequence of SEQ ID NO. 17.
E14: the method of any one of E7 to E13, wherein the CD3 binding domain comprises: a VH comprising the amino acid sequence of SEQ ID NO. 21 and a VL comprising the amino acid sequence of SEQ ID NO. 22.
E15: the method of E13 or E14, wherein the VH and VL of the CD3 binding domain are linked by a linker to form a single chain Fv (scFv).
E16: the method of any one of E13 to E15, wherein the CD3 binding domain comprises the amino acid sequence of SEQ ID No. 23.
E17: the method of any one of E7 to E16, wherein the DLL3 binding domain and the CD3 binding domain are connected by a linker.
E18: the method of any one of E7 to E17, the anti-DLL 3 agent is a bispecific T cell engaging antigen binding polypeptide comprising a DLL3 binding domain and a CD3 binding domain. The DLL3 binding domain comprises (a) a heavy chain variable region (VH) comprising: (i) VH complementarity determining region 1 (CDR-H1) comprising the amino acid sequence of SEQ ID No. 1; (ii) CDR-H2 comprising the amino acid sequence of SEQ ID NO. 2; and (iii) CDR-H3 comprising the amino acid sequence of SEQ ID NO. 3; and (b) a light chain variable region (VL) comprising: (i) VL complementarity determining region 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO. 4; (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 5; and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 6. The CD3 binding domain comprises (a) a VH comprising: (i) a CDR-H1 comprising the amino acid sequence of SEQ ID NO. 18, (ii) a CDR-H2 comprising the amino acid sequence of SEQ ID NO. 19, and (iii) a CDR-H3 comprising the amino acid sequence of SEQ ID NO. 20; and (b) VL comprising: (i) CDR-L1 comprising the amino acid sequence of SEQ ID NO. 15, (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 16, and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 17.
E19: the method of any one of E7 to E18, wherein the DLL3 binding domain comprises a VH comprising the amino acid sequence of SEQ ID No. 7 and a VL comprising the amino acid sequence of SEQ ID No. 8, and wherein the CD3 binding domain comprises a VH comprising the amino acid sequence of SEQ ID No. 21 and a VL comprising the amino acid sequence of SEQ ID No. 22.
E20: the method of any one of E7 to E18, wherein the DLL3 binding domain comprises a VH comprising the amino acid sequence of SEQ ID No. 11 and a VL comprising the amino acid sequence of SEQ ID No. 12, and wherein the CD3 binding domain comprises a VH comprising the amino acid sequence of SEQ ID No. 21 and a VL comprising the amino acid sequence of SEQ ID No. 22.
E21: the method of any one of E7 to E19 wherein the DLL3 binding domain comprises the amino acid of SEQ ID No. 9 and the CD3 binding domain comprises the amino acid of SEQ ID No. 23.
E22: the method of any one of E7 to E18 or E20, the DLL3 binding domain comprises the amino acid of SEQ ID No. 13 and the CD3 binding domain comprises the amino acid of SEQ ID No. 23.
E23: the method of E21 or E22, wherein the anti-DLL 3 agent comprises the amino acid sequence of SEQ ID NO 10 or SEQ ID NO 14.
E24: the method of any one of E7 to E23 wherein the anti-DLL 3 agent comprises the amino acid sequence of SEQ ID No. 27 or SEQ ID No. 32.
E25: the method of any one of E1 to E24, wherein the method further comprises administering one or more additional therapeutic agents to the subject.
E26: the method of E25, wherein the one or more additional therapeutic agents is a corticosteroid (e.g., dexamethasone), saline, or an anti-IL-6 antibody.
E27: the method of any one of E25 or E26, wherein the one or more additional therapeutic agents are administered to the subject in a first cycle in which the anti-DLL 3 agent is administered.
E28: the method of any one of E1 to E27 wherein the anti-DLL 3 agent is prepared by a method wherein a host cell comprising a nucleic acid encoding the anti-DLL 3 agent as described in any one of E7 to E24 is cultured under conditions that allow expression of the anti-DLL 3 agent, and the expressed anti-DLL 3 agent is then recovered from the cell culture.
And E29: the method of any one of E1 to E28, wherein the subject is a human. E30: the method of E29, wherein the subject received at least one prior treatment for the cancer and relapsed, e.g., received two or more prior treatments and relapsed. E31: the method of E30, wherein the at least one prior treatment for the cancer is platinum, chemotherapy, etoposide, and optionally an anti-PD-L1 antibody. E32: the method of E29, wherein the subject has not received prior systemic treatment for the cancer. E33: the method of any one of E1-32, wherein the anti-DLL 3 agent is talastatin. E34: the method of E3, wherein the anti-PD-L1 antibody is alemtuzumab or dewaruzumab.
E33: an anti-DLL 3 agent for treating a DLL3 positive cancer (e.g., SCLC), wherein the anti-DLL 3 agent is administered as described in any one of embodiments E1 to E29.
Drawings
Fig. 1A is a graph showing the optimal percent change from baseline in tumor burden (defined by the sum of the longest diameters (SLD) of all target lesions) for 94 patients whose data expiration date is at least 9 weeks after the first dose date and for whom post-baseline tumor data is available. CR indicates complete remission, PR indicates partial remission, SD indicates stable disease, and NE indicates inaudible. SD indicates that the patient has initial relief but no confirmed relief in subsequent scans, and PR indicates that the patient has initial PR and that future confirmatory scans are still possible. One identified subject in cohort 30 lacks the sum of diameters of lesion measurements and is therefore not included in this figure.Stepped dosing (i.e., 1mg introduced dose) was used in these queues. Fig. 1B is a graph showing the time to respond, duration of treatment, and patient status up to the date of data expiration for the dose of talastatin produced according to all patients with confirmed responses (n=25).
Figure 2A shows Kaplan-Meier curves for progression free survival for patients (n=107) with data expiration dates of at least 9 weeks after the first dosing date. Fig. 2B shows Kaplan-Meier curves for total survival of patients (n=107) with data expiration date of at least 9 weeks after the first dosing date.
FIGS. 3A-3D are graphs showing that peak cytokine levels (6A: IL-6;6B: IL-8;6C: IL-10;6D: TNF- α) tended to be higher in patients with CRS than in patients without CRS. Biomarkers can evaluate patients (n=86); patients with any graded CRS in cycle 1 (n=45); patients not suffering from CRS (n=40). C1, period 1; CRS, cytokine release syndrome; g, grading.
FIG. 4 is a graph showing a longitudinal analysis of IL-10 expression in patients from the phase I study described in example 1. IL-10 showed a significantly higher elevation than the reference normal range and was higher in patients with CRS. JT trend test adjusted P-value = 0.049 (significant at 95% confidence); KW correlation test adjusted P-value = 0.096 (significant at 90% confidence; insignificant at 95% confidence). The yellow dotted line indicates the reference normal range.
FIGS. 5A and 5B are graphs showing analysis of IFN- γ expression in patients from the phase I study described in example 1. IFN-gamma induction is above physiological range; induction was similar between patients with and without cycle 1 CRS. JT trend test adjusted P-value = 0.234 (not significant at 95% confidence); KW correlation test adjusted P-value = 0.317 (not significant at 90% or 95% confidence). The yellow dotted line indicates the reference normal range.
Fig. 6 is a schematic of the clinical study described in example 5.
Detailed Description
AMG 757 was developed for half-life extension for the treatment of DLL3 positive cancers such as SCLC(Bispecific T cell adapter) molecules. The activity of AMG 757 requires simultaneous binding to both target cells (DLL 3 + cells) and T cells. The pharmacological effect of AMG 757 is mediated through specific redirection of previously initiated cytotoxic CD8 + or CD4 + T lymphocytes to kill DLL3 + cells. AMG 757 is being evaluated in the first human study of SCLC subjects (study 20160323) and was found to have anti-tumor activity starting at a dose level of 0.3mg once every two weeks (Q2W) and acceptable safety at doses up to 100mg Q2W.
In preclinical studies, treatment with AMG 757 induced up-regulation of PD-1 and programmed death ligand 1 (PD-L1) on T cells. The combination of AMG 757 and anti-PD-1 antibody increased T cell mediated redirected lysis of DLL3 expressing tumor cells compared to AMG 757 alone (mecianin research report R20190104). Upregulation of PD1/PD-L1 in the tumor microenvironment is a mechanism by which resistance to BiTE therapy is developed, and treatment with anti-PD 1 or anti-PD-L1 therapy may alleviate this resistance.
Combination therapies comprising BiTE molecules and cytotoxic chemotherapy are novel for the treatment of solid tumors. As disclosed and exemplified herein, phase 1 clinical studies using a DLL 3-targeting agent (e.g., AMG 757) alone or in combination with an anti-PD-L1 agent and/or a chemotherapeutic agent to treat SCLC are conducted. The methods disclosed herein may provide improved convenience, flexibility and efficacy and/or reduced side effects to the patient compared to once every two weeks dosing regimens.
1. Definition of the definition
Some of the exemplary bispecific anti-DLL 3 agents disclosed herein (e.g.Molecules) are bispecific T cell engagement antigen binding polypeptides. These polypeptides are recombinant proteins comprising two binding domains, each domain being derived from an antigen-binding fragment of a full-length antibody. Such antigen binding fragments retain the ability to specifically bind antigen (preferably with substantially the same binding affinity). Examples of antigen binding fragments include (i) Fab fragments, monovalent fragments consisting of VL, VH, CL and CH1 domains; (ii) A F (ab') 2 fragment, which is a bivalent fragment comprising two Fab fragments linked at the hinge region by a disulfide bridge; (iii) an Fd fragment consisting of VH and CH1 domains; (iv) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, and (v) dAb fragments (Ward et al, 1989Nature 341:544-546), consisting of the VH domain. Furthermore, although the two domains of the Fv fragment, VL and VH, are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be formed as a single protein chain, in which the VL and VH regions pair to form a monovalent molecule (known as a single chain Fv (scFv); see, e.g., bird et al Science 242:423-426 (1988) and Huston et al, 1988, proc. Natl. Acad. Sci. USA 85:5879-5883.
"Variable domain" refers to the variable region of an antibody light chain (VL) or the variable region of an antibody heavy chain (VH), whether alone or in combination. As is known in the art, the variable regions of the heavy and light chains each consist of four Framework Regions (FR) joined by three Complementarity Determining Regions (CDRs) and contribute to the formation of the antigen binding site of the antibody.
The "complementarity determining regions" (CDRs) of exemplary agents targeting DLL3 are provided in the sequence listing. CDRs may be defined according to the accumulation of Kabat, chothia, kabat and Chothia, abM, contact, north and/or conformational definition or any CDR assay method well known in the art. See, e.g., kabat et al, 1991,Sequences of Proteins of Immunological Interest [ sequence of immunological protein of interest ], 5 th edition (hypervariable regions [ hypervariable region ]); chothia et al, 1989, nature [ Nature ]342:877-883 (structural loop structures [ structural ring structure ]). AbM definition of CDRs is a compromise between Kabat and Chothia and uses AbM antibody modeling software from oxford university molecular Co (Oxfbrd Molecular)The identity of the amino acid residues in a particular antibody constituting a CDR can be determined using methods well known in the art.
The term "treatment" includes prophylactic and/or therapeutic treatment. The treatment is considered prophylactic if it is administered prior to the clinical manifestation of the disorder. Therapeutic treatments include, for example, improving or reducing the severity of a disease, or shortening the length of a disease. Furthermore, the term "treatment" and words related thereto do not necessarily imply 100% or complete treatment. Rather, there are varying degrees of treatment that one of ordinary skill in the art would consider to have potential benefit or therapeutic effect. In this aspect, the cancer treatment methods of the present disclosure can provide any amount or level of treatment. Furthermore, the treatment provided by the methods of the present disclosure may include treating one or more disorders or symptoms or signs of the cancer being treated. Treatment provided by the methods of the present disclosure may also encompass slowing the progression of cancer. For example, these methods can treat cancer by enhancing T cell activity or immune response against cancer, reducing tumor or cancer growth, reducing tumor cell metastasis, increasing cell death of tumor or cancer cells, and the like. In exemplary aspects, the methods treat by delaying the onset or recurrence of cancer for 1 day, 2 days, 4 days, 6 days, 8 days, 10 days, 15 days, 30 days, two months, 4 months, 6 months, 1 year, 2 years, 4 years, or more. In exemplary aspects, the methods are treated by increasing survival of the subject. In various aspects, the treatment provided by the methods of the present disclosure provides a therapeutic response according to a solid tumor remission assessment criteria (RECIST) or other similar criteria. RECIST is a set of criteria established by the united states national cancer institute, canadian national cancer institute clinical trial group, and european cancer research and treatment organization together to assess the progression, stabilization, or responsiveness of tumor and/or cancer cells. According to RECIST, certain tumors are measured at the beginning of an assessment (e.g., clinical trial) to provide a baseline for comparison with post-drug treatment. Eisenhauer et al, eur J Cancer [ J.European J.cancer ]45:228-247 (2009) and Liti re et al, journal of Clinical Oncology [ J.clinical oncology ]37 (13): 1102-1110 (2019) DOI 10.1200/JCO.18.01100 published remission assessment and evaluation criteria for tumors. In various cases, the treatment provided by the methods of the present disclosure provides a therapeutic response assessed in accordance with revised RECIST tumor remission, as follows:
CR = complete remission; na=unavailable; nd=incomplete; PR = partial relief; RECIST = solid tumor remission evaluation criteria; UE = unable to evaluate
a The reduction evaluated relative to baseline. An estimated increase relative to nadir.
b Relief of: for CR and PR, confirmation evaluation is required after not less than 4 weeks, or confirmation evaluation may be performed by waiting for the next predetermined imaging
c Progress: for progressive disease, a confirmatory assessment is required 4 to 6 weeks after the initial imaging progressive disease is observed
d Subjects with only non-indexed lesions
Accordingly, provided herein are methods for: slowing the progression of DLL3 positive cancer in a subject, enhancing T cell activity or immune response in a subject against DLL3 positive cancer, reducing the growth of DLL3 positive tumor or DLL3 positive cancer in a subject, reducing metastasis of DLL3 positive tumor cells in a subject, increasing cell death of DLL3 positive tumor or cancer cells in a subject, delaying onset or recurrence of DLL3 positive cancer in a subject, and/or increasing survival of a subject. In addition, methods of treating DLL3 positive cancers to provide Complete Remission (CR), partial Remission (PR), or Stable Disease (SD) in accordance with revision 1.1 in a subject are also provided. In various aspects, according to the present disclosure, the method comprises administering an anti-DLL 3 agent to a subject alone or in combination with an anti-PD-L1 antibody and/or one or more chemotherapeutic agents. For example, in various aspects, the methods comprise administering an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23, alone or in combination with an anti-PD-L1 antibody and/or a chemotherapeutic agent.
"About" or "approximately" when used in conjunction with a measurable numerical variable refers to the indicated value of the variable and all values of the variable that are within experimental error of the indicated value (e.g., within a 95% confidence interval of the average) or + -10% of the indicated value, whichever is greater. The numerical range includes the numbers defining the range.
"First step dose" or "introduced dose" when used in connection with administration of an anti-DLL 3 agent to treat cancer (e.g., SCLC) refers to the initial dose of the anti-DLL 3 agent in a step dose schedule or regimen. Typically, the first stepped or introduced dose is equal to or lower than the dose at which the first dose effect (e.g., cytokine Release Syndrome (CRS)) is observed. The first step dose may be determined by modeling and simulation of safety and pharmacokinetic data, as is known in the art. For example, the first step dose may be the Maximum Tolerated Dose (MTD) of the anti-DLL 3 agent, where no CRS is observed or CRS below a certain scale (e.g., level 2) is observed.
"Target dose" when referring to administration of an anti-DLL 3 agent for treating cancer (e.g., SCLC) refers to a dose that achieves a target effect of the anti-DLL 3 agent (e.g., improves or reduces the severity of SCLC, or shortens the length of SCLC).
"Step dose" when used in connection with administration of an anti-DLL 3 agent to treat cancer (e.g., SCLC) refers to a dose in a step dose schedule or regimen that is higher than the dose of the previously administered anti-DLL 3 agent. The step dose includes one or more doses that are increased from the first step dose to reach the target dose.
2. DLL3 targeting agents
DLL3 is an atypical Notch ligand that is expressed primarily during the embryogenic process (acting during the somite-generating process). DLL3 accumulates in the Golgi apparatus of normal tissue (Geffers et al, J Cell Biol. [ J. Cell. Biol. ]178:465-476 (2007)). By analyzing the differential expression of DLL3 in 28 SCLC tumors and a large array of normal tissues, DLL3 was identified as a tumor-associated antigen and an attractive target for T cell-based therapies (study 123658).
Human DLL3 proteins contain several extracellular domains: signal peptide, N-terminal, DSL, EGF1, EGF2, EGF3, EGF4, EGF5, EGF6 and membrane proximal domain. The amino acid sequences of human DLL3, EGF3 domain, EGF4 domain, combined EGF3 and EGF4 domain and membrane proximal domain are shown in the sequence listing as SEQ ID NOS 28, 29, 30, 31 and 33, respectively.
Exemplary agents targeting DLL3 are bispecific T cell engaging antigen binding polypeptides that engage DLL3 and CD3, e.gA molecule.The molecule is a recombinant protein made up of two flexibly linked binding domains, each derived from an antibody.One binding domain of the molecule is specific for a tumor associated surface antigen (e.g., DLL 3); the second binding domain is specific for CD3 (a subunit of the T cell receptor complex on T cells). By virtue of its specific design it is possible,The molecules are uniquely suited for transiently linking T cells to target cells and at the same time strongly activate the inherent cytolytic potential of T cells against target cells. See, for example, WO 99/54440, WO 2005/040220 and WO 2008/119567.
Thus, in some embodiments, the described agents targeting DLL3 comprise two binding domains: the first domain binds to DLL3 (preferably human DLL 3) and the second domain binds to CD3 (preferably human CD 3). Preferably, the first domain binds to an epitope of DLL3 comprised within the amino acid sequence of SEQ ID NO. 31. More preferably, the first domain binds to an epitope of DLL3 comprised within the amino acid sequence of SEQ ID NO. 29.
In certain embodiments, the DLL3 binding domain comprises (a) a heavy chain variable region (VH) comprising: (i) VH complementarity determining region 1 (CDR-H1) comprising the amino acid sequence of SEQ ID No. 1; (ii) CDR-H2 comprising the amino acid sequence of SEQ ID NO. 2; and (iii) CDR-H3 comprising the amino acid sequence of SEQ ID NO. 3; and (b) a light chain variable region (VL) comprising: (i) VL complementarity determining region 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO. 4; (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 5; and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 6.
In certain embodiments, the DLL3 binding domain comprises: a VH comprising the amino acid sequence of SEQ ID NO. 7 and a VL comprising the amino acid sequence of SEQ ID NO. 8. In certain preferred embodiments, the DLL3 binding domain comprises: a VH comprising the amino acid sequence of SEQ ID NO. 11 and a VL comprising the amino acid sequence of SEQ ID NO. 12.
In some embodiments, VH and VL are connected by a linker to form a single chain Fv (scFv). In certain embodiments, the DLL3 binding domain comprises the amino acid sequence of SEQ ID NO. 9. In certain preferred embodiments, the DLL3 binding domain comprises the amino acid sequence of SEQ ID NO. 13.
In certain embodiments, the CD3 binding domain comprises: (a) VH comprising: CDR-H1 comprising the amino acid sequence of SEQ ID NO. 18, CDR-H2 comprising the amino acid sequence of SEQ ID NO. 19, and CDR-H3 comprising the amino acid sequence of SEQ ID NO. 20; and VL, comprising: CDR-L1 comprising the amino acid sequence of SEQ ID NO. 15, CDR-L2 comprising the amino acid sequence of SEQ ID NO. 16, and CDR-L3 comprising the amino acid sequence of SEQ ID NO. 17.
In certain embodiments, the CD3 binding domain comprises: a VH comprising the amino acid sequence of SEQ ID NO. 21 and a VL comprising the amino acid sequence of SEQ ID NO. 22. In certain embodiments, the CD3 binding domain comprises the amino acid sequence of SEQ ID NO. 23.
In certain embodiments, an anti-DLL 3 agent disclosed herein comprises two domains. The first domain binds to DLL3 (preferably human DLL 3) and comprises (a) a heavy chain variable region (VH) comprising: (i) VH complementarity determining region 1 (CDR-H1) comprising the amino acid sequence of SEQ ID No. 1; (ii) CDR-H2 comprising the amino acid sequence of SEQ ID NO. 2; and (iii) CDR-H3 comprising the amino acid sequence of SEQ ID NO. 3; and (b) a light chain variable region (VL) comprising: (i) VL complementarity determining region 1 (CDR-L1) comprising the amino acid sequence of SEQ ID NO. 4; (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 5; and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 6. The second domain binds to CD3 (preferably human CD 3) and comprises (a) a VH comprising: (i) a CDR-H1 comprising the amino acid sequence of SEQ ID NO. 18, (ii) a CDR-H2 comprising the amino acid sequence of SEQ ID NO. 19, and (iii) a CDR-H3 comprising the amino acid sequence of SEQ ID NO. 20; and (b) VL comprising: (i) CDR-L1 comprising the amino acid sequence of SEQ ID NO. 15, (ii) CDR-L2 comprising the amino acid sequence of SEQ ID NO. 16, and (iii) CDR-L3 comprising the amino acid sequence of SEQ ID NO. 17.
In certain embodiments, an anti-DLL 3 agent described herein comprises two domains: (a) The first domain binds to DLL3 (preferably human DLL 3) and comprises: a VH comprising the amino acid sequence of SEQ ID NO. 7 and a VL comprising the amino acid sequence of SEQ ID NO. 8; and (b) the second domain binds to CD3 (preferably human CD 3) and comprises: a VH comprising the amino acid sequence of SEQ ID NO. 21 and a VL comprising the amino acid sequence of SEQ ID NO. 22. In certain preferred embodiments, the anti-DLL 3 agents described herein comprise two domains: (a) The first domain binds to DLL3 (preferably human DLL 3) and comprises: a VH comprising the amino acid sequence of SEQ ID NO. 11 and a VL comprising the amino acid sequence of SEQ ID NO. 12; and (b) the second domain binds to CD3 (preferably human CD 3) and comprises: a VH comprising the amino acid sequence of SEQ ID NO. 21 and a VL comprising the amino acid sequence of SEQ ID NO. 22.
In certain embodiments, an anti-DLL 3 agent described herein comprises two domains: (a) The first domain binds to DLL3 (preferably human DLL 3) and comprises the amino acid sequence of SEQ ID NO:9, and (b) the second domain binds to CD3 (preferably human CD 3) and comprises the amino acid sequence of SEQ ID NO: 23. In certain embodiments, an anti-DLL 3 agent described herein comprises two domains: (a) The first domain binds to DLL3 (preferably human DLL 3) and comprises the amino acid sequence of SEQ ID NO:13, and (b) the second domain binds to CD3 (preferably human CD 3) and comprises the amino acid sequence of SEQ ID NO: 23.
In certain embodiments, an anti-DLL 3 agent described herein comprises the amino acid sequence of SEQ ID NO. 10. In certain embodiments, an anti-DLL 3 agent described herein comprises the amino acid sequence of SEQ ID NO: 14.
In certain embodiments, an anti-DLL 3 agent described herein comprises or consists of the amino acid sequence of SEQ ID NO: 27. In certain embodiments, an anti-DLL 3 agent described herein comprises the amino acid sequence of SEQ ID NO: 33.
The anti-DLL 3 agents described herein can be produced by recombinant DNA techniques known in the art. For example, an anti-DLL 3 agent can be produced by a method in which a host cell (e.g., chinese hamster ovary cell) comprising a nucleic acid encoding the anti-DLL 3 agent described herein is cultured under conditions that allow for expression of the anti-DLL 3 agent, and the expressed anti-DLL 3 agent is then recovered from the cell culture. In various embodiments, the anti-DLL 3 agent is Taraxazumab (International non-patent drug name [ INN ]: suggested INN: list 123,WHO Drug Information[WHO drug information ]34 (2): 395-397 (2020)), also known as AMG 757. Taraxazomab is an immunoglobulin scFv-scFv-scFc, anti [ homo sapiens DLL3 (delta-like ligand 3) ] and anti [ homo sapiens CD3E (CD 3 epsilon, leu-4) ], single chain monoclonal antibody (scFv) 2-scFc, bispecific; IG single chain scFv-scFv-scFc, anti-DLL 3 and anti-CD 3E (1-982) [ scFv-VH-V-KAPPA anti-DLL 3 (1-241) [ VH (homo- IGHV4-59*01G49>C(44)(96.9%)-(IGHD)-IGHJ4*01(100%))CDR-IMGT[8.7.12](26-33.51-57.96-107)(1-118)-15-mer tris (tetraglycyl-seryl) linker (119-133) -V-KAPPA (homo- IGKV3-20*01(91.7%)-IGKJ2*01Q120>C(234)(90.9%))CDRIMGT[7.3.9](160-166.184-186.223-231)(134-241)]-6-mer seryl-tetraglycyl-seryl linker (242-247) -scFv-VH-V-LAMBDA anti-CD 3E (248-496) [ VH (mouse IGHV 10-1X 02 (91.9%) - (IGHD) -IGHJ 3X 01 (86.7%)/homo- IGHV3-73*01(87.0%)-(IGHD)-IGHJ5*01(100%))CDR-IMGT[8.10.16](273-280.298-307.346-361)(248-372)-15-mer- tris (tetraglycyl-seryl) linker (373-387) -V-LAMBDA (homo-IGLV 7-43X 01 (85.1%) -IGLJ 3X 02 (100%)) CDR-IMGT [9.3.9] (413-421.439-441.478-486) (388-496-4-mer-tetraglycyl linker (497-500 h) -CH 2-501) (CH 2-CH 2) 3-CH 2-982 ] (CH 3-CH 2-501), nG1m1 (hinge 6-15(501-510),CH2 R83>C(572),N84.4>G(577),V85>C(582)(511-620),CH3 E12(636),M14(638)(621-725),CHS>del)(501-725)-30-mer hexa (tetraglycyl-seryl) linker (726-755) -homo IGHG1 x 03h-CH2-CH3, nG1m1 (hinge 6-15(756-765),CH2 R83>C(827),N84.4>G(832),V85>C(837)(766-875),CH3 E12(891),M14(893)(876-980),CHS(981-982))(756-982)]], non-glycosylated, produced in Chinese Hamster Ovary (CHO) cells; Immunomodulators, anti-tumor properties.
In certain embodiments, exemplary anti-DLL 3 agents are DLL3 (e.g., human DLL 3) binding molecules, such as those disclosed in, for example, WO 2019234220, WO 2020/069028, WO 2019/131988, WO 2021/200898, and WO 2021/155380, all of which are hereby incorporated by reference in their entirety. In certain embodiments, the anti-DLL 3 agent is a protein comprising (a) a first domain that is a single-chain variable fragment that specifically binds to human CD 3; (b) A second domain that is a single domain antibody that specifically binds to human serum albumin; and (c) a third domain that is a single domain antibody that specifically binds to DLL3 protein. In certain embodiments, the anti-DLL 3 agent comprises or consists of the amino acid sequence of SEQ ID NO 34 or 35. In certain embodiments, the anti-DLL 3 agent is a protein comprising (a) a first antigen binding domain that specifically binds to human DLL 3; (b) A second antigen binding domain that specifically binds to human CD3, and (c) first and second Fc domains, wherein the first Fc domain is covalently linked to the first antigen binding domain and the second Fc domain is covalently linked to the second antigen binding domain. In certain embodiments, the first binding domain specifically binds to a membrane proximal region (e.g., SEQ ID NO: 33) of human DLL 3. In certain embodiments, the first binding domain comprises, from N-terminus to C-terminus thereof, a first light chain variable domain, a first light chain constant domain, a first peptide linker, a first heavy chain variable domain, and a first heavy chain constant CH1 domain; and the second binding domain comprises, from its N-terminus to its C-terminus, a second light chain variable domain, a second light chain constant domain, a second peptide linker, a second heavy chain variable domain, and a second heavy chain constant CH1 domain. In certain embodiments, an anti-DLL 3 agent comprises a first antigen binding domain and a second antigen binding domain, wherein the first antigen binding domain comprises or consists of the amino acid sequence of SEQ ID No. 36 and the second binding domain comprises or consists of the amino acid sequence of SEQ ID No. 37. In certain embodiments, the anti-DLL 3 agent comprises a first binding domain and a second binding domain and a third binding domain, wherein at least one of the first and second domains binds to human CD3 and the third binding domain binds to human DLL 3. In certain embodiments, one of the first or second binding domains binds to human CD3, one of the first or second binding domains binds to human CD137, and the third binding domain binds to human DLL 3. In certain embodiments, the first and second binding domains are identical and bind to human CD3, and the third binding domain binds to human DLL 3. In certain embodiments, the first and second binding domains are identical and bind to human CD137, and the third binding domain binds to human DLL 3. Examples of such anti-DLL 3 agents are disclosed in WO 2021200898.
The agents disclosed herein that target DLL3 can be used alone or in combination with other anti-cancer agents disclosed herein to treat DLL3 positive cancers (e.g., lung cancer, small cell lung cancer).
3. Other anticancer agents
A. PD-L1 targeting agents
Programmed cell death protein 1 (PD-1), also known as CD279, SLEB2 and hSLE1, is a transmembrane protein expressed on activated T cells, natural killer cells (NK) and B lymphocytes, macrophages, dendritic Cells (DCs) and monocytes. Notably, PD-1 is highly expressed on tumor-specific T cells (Han et al, am J CANCER RES [ J.Am.cancer research ]10 (3): 727-742 (2020)). PD-1 and B7 protein family members: PD-1 ligand 1 (PD-L1; also known as CD279 and B7-H1) binds to PD-1 ligand 2 (also known as PD-L2, CD273 and B7-DC). PD-L1 is constitutively expressed on T cells and B cells, macrophages and dendritic cells, whereas PD-L2 expression is generally limited to activated DC and macrophages (Xing et al, oncoimmunology [ tumor immunology ]7 (3): e1356144 (2017) (doi: 10.1080/2162402X.2017.1356144)). PD-1 inhibits both adaptive and innate immune responses. The PD-1/PD-L1 axis is associated with the inhibition of T-cell immune responses in cancer. Antagonists of this pathway have been demonstrated clinically in a number of solid tumor indications. PD-1 inhibitors (e.g., nivolumab, pembrolizumab, and cimetidine Li Shan anti [ cemiplimab ]) and PD-L1 inhibitors (e.g., atrazumab, avilamab, and de-valuzumab) target the PD-1/PD-L1 pathway, and each have been approved by the united states Food and Drug Administration (FDA) for the treatment of various cancers. In various embodiments, agents that target PD-L1 (e.g., PD-L1 blockers) can be used in the methods disclosed herein to treat DLL3 positive cancers. Exemplary agents targeting PD-L1 include anti-PD-L1 antibodies, such as alemtuzumab, avilamab, and Dewaruzumab.
In certain embodiments, the anti-PD-L1 antibody is Ab-zumab (International non-patent drug name [ INN ], WHO Drug Information [ WHO drug information ], vol.29, 3 rd, 2015, recommended INN: list 74). Alemtuzumab is a humanized PD-L1 blocking antibody. It is an immunoglobulin G1-kappa, anti [ homo sapiens CD274 (programmed death ligand 1, PDL1, PD-L1, B7 homolog 1, B7H 1) ], humanized monoclonal antibody; gamma 1 heavy chain (1-448) [ humanized VH (homo IGHV3-23 x 04 (86.70%) - (IGHD) -igh hj4 x 01) [8.8.11] (1-118) -homo IGHG1 x 03 (CH 1R 120> K (215) (119-216), hinge (217-231), CH 2N 84.4> a (298) (232-341), CH3 (342-446), CHs (447-448)) (119-448) ], (221-214 ') -disulfide and kappa light chain (1 ' -214 ') [ humanized V-kappa (homo IGKV1-5 x 01 (87.90%) -IGKJ1 x 01) [6.3.9] (1 ' -107 ')) -homo IGKC 01 (108 ' -214 ') ]; dimer (227-227 ': 230-230') -bis-disulfide. Alemtuzumab is commercially available, e.g., asAnd (5) marketing.
In certain embodiments, the anti-PD-L1 antibody is avermectin (International non-patent drug name [ INN ], WHO Drug Information [ WHO drug information ], vol.30, stage 1, 2016, INN recommended: list 75). Avermectin is a PD-L1 blocking monoclonal antibody produced in CHO cells. It is an immunoglobulin G1- λ1, anti [ homo sapiens CD274 (programmed death ligand 1, PDL1, PD-L1, B7 homolog 1, B7H 1) ], homo sapiens monoclonal antibody; gamma 1 heavy chain (1-450) [ homo sapiens VH (IGHV 3-23 x 01 (90.80%) - (IGHD) -IGHJ4 x 01) [8.8.13] (1-120) -IGHG 1x 01, gm17,1 (CH 1 (121-218), hinge (219-233), CH2 (234-343), CH3 (344-448), CHs (449-450) (121-450) ], (223-215 ') -disulfide and λ1 light chain (1 ' -216 ') [ homo sapiens V-LAMBDA (IGLV 2-14 x 01 (99.00%) -IGLJ 1x 01) [9.3.10] (1 ' -110 ') -aba IGLC 1x 02 (111 ' -216 '); dimer (229-229 ": 232-232") -disulfide monoclonal antibody is commercially available, for example, asAnd (5) marketing.
In certain embodiments, the anti-PD-L1 antibody is Dewaruzumab (International non-patent drug name [ INN ], WHO Drug Information [ WHO drug information ], vol.29, 3, 2015, recommended INN: list 74). Dewaruzumab is a PD-L1 blocking monoclonal antibody produced in CHO cells. It is an immunoglobulin G1- κ, anti [ homo sapiens CD274 (programmed death ligand 1, pdl1, pd-L1, B7 homolog 1, B7H 1) ], homo sapiens monoclonal antibody; gamma 1 heavy chain (1-451) [ homo VH (IGHV 3-7X 01 (99.00%) - (IGHD) -IGHJ 4X 01) [8.8.14] (1-121) -IGHG 1X 03 (CH 1 (122-219), hinge (220-234), CH2 (235-344) L1.3>F (238),L1.2>E(239),P116>S(335),CH3(345-449),CHS(450-451))(122-451)],(224-215')- disulfide and KAPPA light chain (1 '-215') [ homo V-KAPPA (IGKV 3-20X 01 (96.90%) -IGKJ 1X 01) [7.3.9] (1 '-108') -IGKC X01 (109 '-215') ]; dimer (230-230 ": 233-233") -double disulfide Dewarukab commercially available, for example, asAnd (5) marketing.
B. Chemotherapeutic agents
"Chemotherapeutic agents" are also referred to as antineoplastic agents, and include compounds useful in the treatment of cancer. Chemotherapeutic agents may be classified according to their mechanism of action and may be further divided into subgroups within each class. Exemplary classes of chemotherapeutic agents include alkylating agents, antimetabolites, topoisomerase inhibitors, antitumor antibiotics, mitotic inhibitors, and protein kinase inhibitors. Alkylating agents include subgroups such as oxazaphosphorine (oxazaphosphorine), nitrogen mustard, imidazotetrazine, nitrosoureas, alkyl sulfonates, hydrazines, and platinum-based agents. Platinum-based agents include cisplatin, carboplatin, and oxaliplatin. Topoisomerase inhibitors include topoisomerase I inhibitors and topoisomerase II inhibitors. Mitotic inhibitors include vinca alkaloids, taxanes and non-taxane microtubule inhibitors. Antitumor antibiotics include bleomycin, actinomycin D (dactinomycin) and mitomycin. Protein kinase inhibitors include BCR-ABL and c-KIT tyrosine kinase inhibitors, EGFR tyrosine kinase inhibitors, ALK tyrosine kinase inhibitors, V600E mutant BRAF oncogene inhibitors, MEK inhibitors, bruton kinase inhibitors, janus kinase inhibitors, and CDK inhibitors.
In certain embodiments, the chemotherapeutic agent useful in the methods disclosed herein is an alkylating agent. In certain embodiments, the alkylating agent is a platinum-based agent, such as cisplatin, carboplatin, or oxaliplatin. In certain embodiments, the alkylating agent is lubidine (lurbinectedin). The lubitidine is commercially available, for example, as Zepzelca TM. In certain embodiments, the chemotherapeutic agents useful in the methods disclosed herein are topoisomerase inhibitors, e.g., topoisomerase II inhibitors (e.g., etoposide). In certain embodiments, chemotherapeutic agents useful in the methods disclosed herein include platinum-based drugs (cisplatin, carboplatin, or oxaliplatin), topoisomerase II inhibitors (etoposide), or a combination of platinum-based agents and topoisomerase II inhibitors.
4. Dosing regimen for DLL 3-targeted cancer treatment
Disclosed herein are methods of treating DLL3 positive cancers (e.g., lung cancer, SCLC, neuroendocrine prostate cancer [ NEPC ]) comprising administering to a subject in need thereof an agent that targets DLL3, or a combination of agents and/or chemotherapeutic agents that target DLL3 and PD-L1. The method comprises administering an agent that targets DLL3 alone or in combination with an agent that targets PD-L1 and/or a chemotherapeutic agent to a subject in need thereof according to the specific dosages/regimens disclosed herein. In certain embodiments, the method further comprises administering to the subject one or more additional therapeutic agents that prevent, reduce, or mitigate the risk of side effects associated with administration of the DLL3 targeted agent. In various embodiments, the agent that targets DLL3 is administered by parenteral administration. In various embodiments, the DLL 3-targeting agent is administered by Intravenous (IV) infusion. The DLL 3-targeting agent is administered by bolus IV infusion (e.g., infusion over about 60 minutes) unless otherwise indicated herein. Similarly, agents targeting PD-L1 are administered by bolus IV infusion unless otherwise indicated.
A. dosing regimen using DLL3 targeted agents
In one aspect, disclosed herein are methods of treating DLL3 positive cancers comprising administering to a subject in need thereof an agent that targets DLL 3. Agents targeting DLL3 include the anti-DLL 3 agents disclosed above. In certain embodiments, disclosed herein is a method of treating a DLL3 positive cancer, the method comprising administering an anti-DLL 3 agent to a subject in need thereof at a dose of from about 3mg to about 100mg twice every three weeks (21 days). In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 10mg to about 100mg twice every three weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of about 3mg, about 10mg, about 30mg, 50mg, about 80mg, or about 100mg, particularly about 10mg, about 30mg, or about 100mg twice every three weeks. In certain embodiments, the anti-DLL 3 agent is administered on days 1 and 8 of a 21 day cycle.
In certain embodiments, disclosed herein is a method of treating a DLL3 positive cancer, the method comprising administering an anti-DLL 3 agent to a subject in need thereof at a dose of from about 6mg to about 200mg once every three weeks (21 days, Q3W). In certain embodiments, the anti-DLL 3 agent is administered at a dose in the range of from about 6mg to about 50mg, or about 20mg to about 100mg, or from about 80mg to about 150mg, or from about 100mg to about 200mg once every three weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of about 6mg, about 20mg, about 60mg, about 100mg, about 180mg, or about 200mg, particularly about 20mg, about 60mg, or about 200mg once every three weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of about 20mg once every three weeks. In certain embodiments, the anti-DLL 3 agent is administered on day 1 of a 21 day cycle.
Due to the mechanism of action of anti-DLL 3 agents (e.g., DLL 3-targeted agents disclosed above), during initial treatment with anti-DLL 3 agents, subjects may have an increased risk of Cytokine Release Syndrome (CRS), and a step administration method may be implemented. Thus, in certain embodiments, the anti-DLL 3 agent is administered during the beginning of the agent treatment (e.g., the first cycle of treatment) using a step dosing method, after which the anti-DLL 3 agent is administered according to the above-described twice-every-three-week or once-every-three-week regimen. In such embodiments, anti-DLL 3 is administered during the beginning of treatment (cycle 1) at 21 day periods according to the following step dose regimen: a first step dose or lead-in dose on day 1, a step dose equal to the target dose on day 8, and a target dose on day 15. Alternatively, the anti-DLL 3 agent is administered during the beginning of treatment (cycle 1) at 21 day periods according to the following: a first dose or an introduced dose is administered on day 1, a stepped dose equal to the target dose is administered on day 8, and no anti-DLL 3 agent is administered on day 15; or no anti-DLL 3 agent on day 1, a first dose or lead-in dose on day 8, and a step dose equal to the target dose on day 15.
In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 0mg or 1mg on day 1, a second dose on day 8, and a third dose on day 15, b) one or more subsequent doses administered twice per cycle beginning on cycle 2 of the 21 day cycle and thereafter, wherein each of the second, third, and one or more subsequent doses is the same and is from about 3mg to about 100mg. In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 1mg on day 1, a second dose on day 8, and a third dose on day 15, b) one or more subsequent doses administered twice per cycle beginning on cycle 2 of the 21-day cycle and thereafter, wherein each of the second, third, and one or more subsequent doses is the same and is from about 10mg to about 100mg (e.g., 10mg, 30mg, or 100 mg). In certain embodiments, the anti-DLL 3 agent is administered at cycle 2 and thereafter at days 1 and 8.
In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 0mg or 1mg on day 1, a second dose on day 8, and a third dose on day 15, b) one or more subsequent doses administered once per cycle beginning on cycle 2 of the 21 day cycle and thereafter, wherein each of the second dose, third dose, and one or more subsequent doses is the same and is from about 6mg to about 200mg. In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 1mg on day 1, a second dose on day 8, and a third dose on day 15, b) one or more subsequent doses administered once per cycle beginning on cycle 2 of the 21 day cycle and thereafter, wherein each of the second dose, the third week dose, and the one or more subsequent doses is the same and is from about 20mg to about 200mg (e.g., 20mg, 60mg, or 200 mg). In certain embodiments, the anti-DLL 3 agent is administered at cycle 2 and thereafter at day 1.
In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 1mg on day 1, a second dose on day 8, b) one or more subsequent doses administered once per cycle beginning on cycle 2 of the 21-day cycle and thereafter, wherein the second dose is the same as each of the one or more subsequent doses and is from about 6mg to about 100mg (e.g., 20 mg). In such embodiments, no anti-DLL 3 agent is administered on day 15 of cycle 1.
In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Cycle 1: a first dose of 1mg on day 8, a second dose on day 15, b) one or more subsequent doses administered once per cycle beginning on cycle 2 of the 21-day cycle and thereafter, wherein the second dose is the same as each of the one or more subsequent doses and is from about 6mg to about 100mg (e.g., 20 mg). In such embodiments, no anti-DLL 3 agent is administered on day 1 of cycle 1.
In the various embodiments described above, the anti-DLL 3 agent is administered by parenteral administration, such as bolus IV infusion (e.g., infusion over about 60 minutes). The twice-every-three-week and once-every-three-week dosing regimen described above can provide improved convenience and flexibility to the patient and can increase the activity of the anti-DLL 3 agent as compared to twice-every-two-week dosing regimen.
In addition to the step dosing method, during the initiation of the agent treatment (e.g., cycle 1 of treatment), the anti-DLL 3 agent may be administered via IV infusion over an extended period of time (eIV) (e.g., over a period of 2 to 7 days). eIV can achieve similar or higher anti-DLL 3 agent cumulative serum exposure and have a lower Cmax compared to bolus IV administration (e.g., IV infusion over about 60 minutes). Thus, use eIV may reduce the intensity and/or frequency of symptoms associated with CRS while achieving similar or enhanced pharmacodynamic activity (e.g., efficacy). eIV are also referred to herein as continuous intravenous infusion.
In certain embodiments, the anti-DLL 3 agent is administered during the beginning of the agent treatment (first cycle of treatment) using the eIV method, after which the anti-DLL 3 agent is administered according to the two-week or once-every-three-week regimen described above. In such embodiments, anti-DLL 3 is administered at a 21 day period, wherein the anti-DLL 3 agent is administered at cycle 1 according to the following: starting on day 1, administration is by continuous intravenous infusion at a dose of from about 1mg to about 200mg over a period of 2 days to 7 days, and optionally, intravenous infusion by bolus injection at a dose of from about 10mg to about 200mg on day 8, day 15, or both days 8 and 15.
In certain embodiments, the anti-DLL 3 agent is administered at 21 day periods according to the following schedule: a) Beginning on day 1 of cycle 1, administering an anti-DLL 3 agent by continuous intravenous infusion at a dose of from about 1mg to about 200mg over a period of 2 days to 7 days, and b) beginning on cycle 2 and thereafter, administering an anti-DLL 3 agent by bolus intravenous infusion as follows: i) Twice every three weeks at a dose of from about 10mg to about 100mg, or ii) once every three weeks at a dose of from about 20mg to about 200 mg. In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 10mg to about 100mg or from about 30mg to about 100mg over a period of 2 days, 3 days, 5 days, or 7 days in a). In certain embodiments, the anti-DLL 3 agent is administered at a dose from about 100mg to about 200mg over a period of 2 days, 3 days, 5 days, or 7 days in a). In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 30mg, 50mg, or 100mg over a period of 3 days, 5 days, or 7 days in a). In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 30mg or 100mg over a period of 3 days in a). In certain embodiments, the method further comprises a) administering the anti-DLL 3 agent by bolus IV infusion at a dose of from about 10mg to about 200mg on day 8, day 15, or both days 8 and day 15 of cycle 1.
In certain embodiments, the anti-DLL 3 agent is administered at a 28 day period according to the following regimen: a) Starting on day 1 of cycle 1, administering an anti-DLL 3 agent by continuous intravenous infusion at a dose of from about 1mg to about 100mg over a period of 2 days to 7 days, and b) starting on cycle 2 and thereafter, administering an anti-DLL 3 agent by bolus intravenous infusion at a dose of from about 10mg to about 100mg once every two weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 10mg to about 100mg or from about 30mg to about 100mg over a period of 2 days, 3 days, 5 days, or 7 days in a). In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 30mg, 50mg, or 100mg over a period of 3 days, 5 days, or 7 days in a). In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 30mg or 100mg over a period of 3 days in a). In certain embodiments, the method further comprises a) administering the anti-DLL 3 agent by bolus IV infusion at a dose of from about 10mg to about 100mg on day 8, day 15, or both days 8 and day 15 of cycle 1.
Anti-DLL 3 agents include any of the DLL3 targeted agents described above. For example, an anti-DLL 3 agent comprises or consists of the amino acid sequences of SEQ ID NOs 13 and 23, or comprises or consists of the amino acid sequences of SEQ ID NOs 14, 27 or 32. In various embodiments, the anti-DLL 3 agent is AMG 757. In various embodiments, the DLL3 positive cancer comprises lung cancer, such as SCLC or NEPC. In certain embodiments, the SCLC is recurrent/refractory SCLC (RR SCLC) or extensive disease SCLC (ED SCLC), or restricted stage SCLC. In certain embodiments, the subject is a human having SCLC (e.g., RR SCLC or ED SCLC).
B. Dosing regimen using agents targeting DLL3 and PD-L1
Disclosed herein are methods of treating DLL3 positive cancers comprising administering to a subject in need thereof a combination of agents that target DLL3 and PD-L1. Agents targeting DLL3 include anti-DLL 3 agents disclosed herein, and agents targeting PD-L1 include anti-PD-L1 antibodies disclosed herein. In one aspect, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered once every two weeks at a dose of from about 10mg to about 100 mg. In certain embodiments, the anti-DLL 3 agent is administered at a dose of about 10mg, about 30mg, about 50mg, or about 100mg once every two weeks. In certain embodiments, the anti-DLL 3 agent is administered on days 1 and 15 of a 28 day cycle.
In one aspect, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered twice every three weeks at a dose of from about 10mg to about 100mg or once every three weeks at a dose of from about 20mg to about 200 mg. In certain embodiments, the anti-DLL 3 agent is administered, for example, twice every three weeks at a dose of about 10mg, about 30mg, about 50mg, or about 100mg on days 1 and 8 of a 21 day cycle. In certain embodiments, an anti-DLL 3 agent is administered, for example, once every three weeks at a dose of from about 20mg to about 100mg (e.g., about 20mg, about 60mg, or about 100 mg) on day 1 of a 21 day cycle. In certain embodiments, an anti-DLL 3 agent is administered, for example, once every three weeks at a dose of from about 100mg to about 200mg (e.g., about 120mg or about 200 mg) on day 1 of a 21 day cycle.
In various embodiments in which an anti-DLL 3 agent is administered with an anti-PD-L1 antibody and optionally one or more chemotherapeutic agents, the anti-DLL 3 agent may be administered according to a step dose regimen during the beginning of treatment (e.g., cycle 1) to minimize potential side effects (e.g., CRS) associated with the anti-DLL 3 agent. Thus, in certain embodiments, the anti-DLL 3 agent is administered in a21 day period according to the following regimen during cycle 1 of treatment: a first dose of about 0mg or about 1mg on day 1, a second dose of from about 1mg to about 100mg on day 8, and a third dose of from about 10mg to about 200mg on day 15. In certain embodiments, the anti-DLL 3 agent is administered in cycle 1 according to the following regimen: a first dose of about 1mg on day 1, a second dose of from about 10mg to about 100mg on day 8, and a third dose of from about 10mg to about 100mg on day 15. In certain embodiments, the anti-DLL 3 agent is administered in cycle 1 according to the following regimen: a first dose of about 1mg on day 1, a second dose of from about 10mg to about 100mg on day 8, and a third dose of from about 20mg to about 200mg on day 15. Each cycle 1 regimen may be used prior to the two week, or three week regimen of the anti-DLL 3 agent described above.
In certain preferred embodiments, wherein the anti-DLL 3 agent is administered in combination with the anti-PD-L1 agent and optional chemotherapy, the anti-DLL 3 agent is administered in a 21 day cycle in treatment cycle 1 according to the following regimen: a first dose of about 1mg is administered on day 1, a second dose (e.g., 20 mg) from about 10mg to about 100mg is administered on day 8, and no anti-DLL 3 agent is administered on day 15. Alternatively, the anti-DLL 3 agent is administered in a 21 day period according to the following regimen in period 1 of treatment: no anti-DLL 3 agent is administered on day 1, a first dose of about 1mg is administered on day 8, and a second dose (e.g., 20 mg) from about 10mg to about 100mg is administered on day 15. In such embodiments, if administered on the same day, the anti-PD-L1 agent (de-valuzumab or alemtuzumab) is administered first, followed by administration of one or more chemotherapeutic agents and the anti-DLL 3 agent.
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered according to the following regimen: a) Cycle 1 (21 days): about 1mg first dose on day 1, second dose on day 8, third dose on day 15, b) cycle 2 and cycle 3 (21 days per cycle): a fourth dose on day 1 and a fifth dose on day 8 of each cycle, and c) one or more subsequent doses beginning on day 4 and once every two weeks thereafter at a 28-day cycle, wherein the second dose, the third dose, the fourth dose, the fifth dose, and the one or more subsequent doses are the same and are each from about 10mg to about 100mg (e.g., about 10mg, about 30mg, or about 100 mg).
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered at a 21 day period according to the following schedule: a) Cycle 1: about 1mg first dose on day 1, second dose on day 8, third dose on day 15, b) cycle 2 and 3: a fourth dose on day 1 and a fifth dose on day 8 of each cycle, and c) one or more subsequent doses beginning at cycle 4 and once every three weeks thereafter, wherein the second dose, the third dose, the fourth dose, and the fifth dose are the same and are each from about 10mg to about 100mg (e.g., about 10mg, about 30mg, or about 100 mg), and wherein the one or more subsequent doses are the same and are each from about 20mg to about 200mg (e.g., about 20mg, about 60mg, or about 200 mg).
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered at a 21 day period according to the following schedule: a) Cycle 1: a first dose of about 1mg on day 1, a second dose of from about 10mg to about 100mg on day 8, a third dose on day 15, b) cycle 2 and cycle 3: a fourth dose on day 1 of each cycle, and c) one or more subsequent doses beginning at cycle 4 and once every three weeks thereafter, wherein the third dose, the fourth dose, and the one or more subsequent doses are the same and are each from about 20mg to about 200mg (e.g., about 20mg, about 60mg, or about 200 mg).
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered at a 21 day period according to the following schedule: a) Cycle 1: a first dose of about 1mg on day 1, a second dose (e.g., 20 mg) from about 10mg to about 100mg on day 8, b) cycle 2 and cycle 3: a third dose on day 1 of each cycle, and c) one or more subsequent doses beginning at cycle 4 and once every three weeks thereafter, wherein the third dose is the same as the one or more subsequent doses and is each from about 10mg to about 100mg (e.g., about 20 mg).
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered at a 21 day period according to the following schedule: a) Cycle 1: a first dose of about 1mg on day 8, a second dose (e.g., 20 mg) from about 10mg to about 100mg on day 15, b) cycle 2 and cycle 3: a third dose on day1 of each cycle, and c) one or more subsequent doses beginning at cycle 4 and once every three weeks thereafter, wherein the third dose is the same as the one or more subsequent doses and is each from about 10mg to about 100mg (e.g., about 20 mg).
In various embodiments wherein the anti-DLL 3 agent is administered with an anti-PD-L1 antibody and optionally one or more chemotherapeutic agents, the anti-PD-L1 antibody is a PD-L1 blocking antibody. Examples of such anti-PD-L1 antibodies include alemtuzumab, dewaruzumab, and avilamab. In certain embodiments, the anti-PD-L1 antibody is alemtuzumab or dewaruzumab. In certain embodiments, the anti-PD-L1 antibody is Dewaruzumab. When used in the methods disclosed herein, the dosage and regimen of the anti-PD-L1 antibodies are the same as approved by regulatory authorities (e.g., FDA). For example, alemtuzumab can be administered at a dose of about 840mg every 2 weeks, or about 1200mg every 3 weeks, or about 1680mg every 4 weeks. For example, dewaruzumab may be administered at a dose of about 10mg/kg every 2 weeks, or about 1500mg every 3 weeks, or about 1500mg every 4 weeks.
In various embodiments wherein the anti-DLL 3 agent is administered with an anti-PD-L1 antibody and optionally one or more chemotherapeutic agents, the one or more chemotherapeutic agents comprise an alkylating agent, a topoisomerase inhibitor, or a combination thereof. In certain embodiments, the one or more chemotherapeutic agents include a platinum-based agent (e.g., cisplatin, carboplatin, or oxaliplatin), a topoisomerase II inhibitor (e.g., etoposide), or a combination thereof. In certain embodiments, the one or more chemotherapeutic agents comprise cisplatin or carboplatin and etoposide. In certain embodiments, the one or more chemotherapeutic agents is etoposide. In various embodiments, one or more chemotherapeutic agents are administered according to a regulatory body (e.g., FDA) approved dosage and/or regimen. For example, in various embodiments, carboplatin is administered at a dose sufficient to achieve AUC = 5mg/ml/min, and etoposide is administered at a dose of 100mg/m 2.
Anti-DLL 3 agents include any of the DLL3 targeted agents described above. For example, an anti-DLL 3 agent comprises or consists of the amino acid sequences of SEQ ID NOs 13 and 23, or comprises or consists of the amino acid sequences of SEQ ID NOs 14, 27 or 32. In various embodiments, the DLL3 positive cancer comprises lung cancer, such as SCLC or NEPC. In certain embodiments, the SCLC is recurrent/refractory SCLC (RR SCLC) or extensive disease SCLC (ED SCLC) or restricted stage SCLC. In certain embodiments, the subject is a human having SCLC (e.g., RR SCLC, ED SCLC, or restricted SCLC).
In various embodiments, the anti-DLL 3 agent, the anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents are each administered by IV infusion. In certain embodiments, the anti-DLL 3 agent is administered after administration of the anti-PD-L1 antibody and the one or more chemotherapeutic agents when administered on the same day.
In various embodiments disclosed herein, "combination therapy" or "in combination with … …" refers to the administration of one therapeutic modality (e.g., an anti-DLL 3 agent) in addition to another therapeutic modality (e.g., an anti-PD-L1 antibody and optionally one or more chemotherapeutic agents) to a subject (e.g., a human) having a DLL3 positive cancer. In combination therapies in which an anti-DLL 3 agent and an anti-PD-L1 antibody are involved, one treatment modality may be administered before, during, or after another treatment modality is administered to a subject. Such combination therapies do not include situations in which 28 days or more have elapsed between the end of administration of one treatment modality and the beginning of administration of another treatment modality.
C. Dosing regimen with anti-DLL 3 agents and chemotherapeutic agents
Disclosed herein are methods of treating DLL3 positive cancers comprising administering to a subject in need thereof an anti-DLL 3 agent in combination with one or more chemotherapeutic agents. Agents that target DLL3 include anti-DLL 3 agents disclosed herein, and chemotherapeutic agents include alkylating agents disclosed herein. In certain embodiments, the alkylating agent is lubidine.
In certain embodiments, disclosed herein is a method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent and an alkylating agent, wherein the anti-DLL 3 agent is administered to the subject at a dose of from about 10mg to about 200mg (e.g., 10mg, 20mg, 60mg, or 100 mg) once every two weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 10mg to about 100mg (e.g., 10mg, 20mg, 60mg, or 100 mg) twice every three weeks. In certain embodiments, the anti-DLL 3 agent is administered at a dose of from about 20mg to about 200mg (e.g., 20mg, 60mg, 100mg, or 200 mg) once every three weeks. In certain embodiments, the anti-DLL 3 agent is administered on day 1 of a 21 day cycle. In various embodiments, the alkylating agent is lubidine.
In certain embodiments, the method comprises administering to a subject in need thereof an anti-DLL 3 agent and an alkylating agent, wherein the anti-DLL 3 agent is administered to the subject at a 21 day period according to: a first dose of 0mg or about 1mg on day 1, a second dose of from about 10mg to about 100mg on day 8, a third dose of from about 10mg to about 200mg on day 15, and one or more subsequent doses of from about 10mg to about 200mg beginning on day 22 and once every three weeks thereafter. In certain embodiments, the first dose is 1mg, the second dose is from 10mg to 100mg (e.g., 10mg, 20mg, 60mg, or 100 mg), the third dose is from 10mg to 200mg (e.g., 10mg, 20mg, 60mg, 100mg, or 200 mg), and one or more subsequent doses are the same and the same as the third dose (e.g., 10mg, 20mg, 60mg, 100mg, or 200 mg). In certain embodiments, the method comprises administering only the alkylating agent in cycle 1 and cycle 2, and administering the alkylating agent and the anti-DLL 3 agent in cycle 3 and thereafter. In various embodiments, the alkylating agent is lubidine. In various embodiments, the lubidine may be administered according to a regulatory body (e.g., FDA) approved dose and/or regimen, e.g., once every three weeks at a dose of about 3.2mg/m 2、2.6mg/m2 or 2mg/m 2.
Anti-DLL 3 agents include any of the DLL3 targeted agents described above. For example, an anti-DLL 3 agent comprises or consists of the amino acid sequences of SEQ ID NOs 13 and 23, or comprises or consists of the amino acid sequences of SEQ ID NOs 14, 27 or 32
In various embodiments, the DLL3 positive cancer includes lung cancer, such as SCLC. In certain embodiments, SCLC is recurrent/refractory SCLC (RR SCLC) or extensive disease SCLC (ED SCLC). In certain embodiments, the subject is a human having SCLC (e.g., RR SCLC or ED SCLC).
In various embodiments, the anti-DLL 3 agent and the alkylating agent (e.g., lubidine) are each administered by IV infusion. In certain embodiments, the anti-DLL 3 agent is administered after administration of the alkylating agent when administered on the same day.
In various embodiments disclosed herein, "combination therapy" or "in combination with … …" refers to the administration of one therapeutic modality (e.g., an anti-DLL 3 agent) in addition to another therapeutic modality (e.g., an alkylating agent, such as lubidine) to a subject (e.g., a human) having a DLL3 positive cancer. In combination therapies in which an anti-DLL 3 agent and lubidine are involved, one treatment regimen may be administered before, during, or after another treatment regimen is administered to the subject. Such combination therapies do not include situations in which 28 days or more have elapsed between the end of administration of one treatment modality and the beginning of administration of another treatment modality.
5. Additional therapeutic agents
In some embodiments, the methods disclosed herein further comprise using one or more additional therapeutic agents to prevent, reduce, or mitigate the risk of side effects associated with administration of an anti-DLL 3 agent alone or in combination with an anti-PD-L1 antibody and/or chemotherapeutic agent. The primary side effect associated with the use of anti-DLL 3 agents is CRS. One or more additional therapeutic agents useful for preventing, reducing, or alleviating the risk of CRS include corticosteroids (e.g., dexamethasone), liquids (e.g., saline), and anti-IL 6 antibodies (e.g., tolizumab or steuximab [ siltuximab ]). Dexamethasone can be administered IV prior to all cycle 1 doses (including all step doses) of the anti-DLL 3 agent (e.g., AMG 757), saline (e.g., 1 liter) can be administered IV after all anti-DLL 3 agent (e.g., AMG 757) doses in cycle 1, and an anti-IL 6 antibody (e.g., tolizumab or steuximab) can be administered as needed (e.g., subject is non-responsive to IV fluid). Additional corticosteroid prophylaxis may be achieved with oral dexamethasone as desired. Exemplary doses of dexamethasone include 8 mg/administration (maximum 24 mg/day). Exemplary doses of tolizumab include 8mg/kg (not more than 800 mg). Symptoms of CRS include fever, nausea, fatigue, headache, myalgia, weakness, and therapeutic agents useful for treating these symptoms (e.g., acetaminophen/acetaminophen for fever) may also be used. In certain embodiments, one or more additional therapeutic agents that may also be used to reduce or mitigate side effects associated with anti-DLL 3 agent treatment include granulocyte colony stimulating factor (e.g., febuxostat or pefebuxostat).
Thus, in certain embodiments, the methods disclosed herein further comprise administering one or more additional therapeutic agents selected from the group consisting of corticosteroids (e.g., prednisone, hydrocortisone, and dexamethasone), liquids (saline), and anti-IL 6 antibodies (e.g., tolizumab or stetuximab). In certain embodiments, the methods further comprise administering one or more additional therapeutic agents selected from the group consisting of corticosteroids (e.g., dexamethasone), liquids (saline), and tolizumab or cetuximab. In certain embodiments, one or more of a corticosteroid, a liquid, and tolizumab are administered in cycle 1, where an anti-DLL 3 agent (e.g., AMG 757) is administered.
In certain embodiments of any one of the methods in which one or more additional therapeutic agents are administered, the subject is a human.
6. Therapeutic response
The efficacy of the disclosed methods for treating DLL3 positive cancers (e.g., SCLC) can be assessed by a variety of clinical outcomes, endpoints, and/or metrics. In this regard, the clinical outcomes that can be assessed include, but are not limited to, progression Free Survival (PFS), total survival (OS), objective remission (ORR), disease Control Rate (DCR), duration of remission (DOR). As used herein, the term "Progression Free Survival (PFS)" refers to the time from randomization until the first evidence of disease progression or death. As used herein, the term "total lifetime (OS)" refers to the time from random to death. As used herein, the term "Objective Remission Rate (ORR)" is a measure of how a particular treatment affects tumor burden in a patient with a history of solid tumors, and refers to the proportion of patients that produce partial or complete remission to therapy. As used herein, the term "duration of remission (DoR)" refers to the time from randomness to disease progression or death in a patient that achieves complete or partial remission. As used herein, the term "Disease Control Rate (DCR)" describes the percentage of advanced cancer patients whose therapeutic intervention results in complete remission, partial remission, or stable disease. Clinical endpoints for cancer therapies are further described, for example, in Delgado A. And Guddati, A.K. et al, am J CANCER RES [ J.S. J.cancer research ]2021;11 (4) 1121-1131.
In some aspects, the disclosed methods contemplate increasing one or more of Progression Free Survival (PFS), total survival (OS), objective Remission Rate (ORR), and/or Disease Control Rate (DCR) and duration of remission (DOR) as compared to standard of care (SOC). For example, when the anti-DLL 3 agent is used as a first line treatment for SCLC (e.g., the anti-DLL 3 agent treats a patient not previously systemically treated in combination with anti-PD L1 and optionally a chemotherapeutic agent), the disclosed methods are expected to cause an ORR of at least about 65% (e.g., about 65% to about 70%), a median PFS of greater than about 5 months (e.g., 7 months or more), and a median OS of at least about 13 months (e.g., about 13 to 15 months or about 16 to 18 months). When the anti-DLL 3 agent is administered as a tri-line treatment of SCLC (e.g., anti-DLL 3 agent monotherapy treatment of a patient who has received two or more prior therapies and relapsed), the method is expected to cause an ORR of at least about 20% (e.g., about 24% or more), a median PFS of greater than about 6 months, and a median OS of at least about 7.5 months (e.g., 8 months or more).
7. Article of manufacture
Disclosed herein are articles comprising: (a) a container containing an anti-DLL 3 agent (e.g., AMG 757); and (b) a package insert carrying instructions for treating DLL3 positive cancer (or treating SCLC or NEPC) in a subject by administering an anti-DLL 3 agent (e.g., AMG 757), wherein the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose from about 10mg to about 100mg (e.g., 10mg, 30mg, or 100 mg) once every two weeks (e.g., on days 1 and 8 of a 21 day cycle). In certain embodiments, the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose of from about 20mg to about 200mg (e.g., 20mg, 60mg, 100mg, 160mg, or 200 mg) once every three weeks (e.g., on day 1 of a 21 day cycle). In certain embodiments, the instructions further specify that in a first cycle of administering an anti-DLL 3 agent to a subject, the anti-DLL 3 agent is administered by prolonged intravenous infusion over 2 days to 7 days (e.g., over 3 days). In certain embodiments, the instructions further specify that the anti-DLL 3 agent is to be administered according to a step dosing regimen in cycle 1 (21-day cycle): 1mg on day 1, a second dose on day 8, and a third dose on day 15, wherein the second and third doses are the same and are each from about 10mg to about 100mg.
In certain embodiments, an article of manufacture comprises: (a) a container containing an anti-DLL 3 agent (e.g., AMG 757); and (b) a package insert carrying instructions for treating a DLL3 positive cancer (or treating SCLC) in a subject by administering an anti-DLL 3 agent (e.g., AMG 757) in combination with an anti-PD-L1 antibody (e.g., alemtuzumab or de-valuzumab), wherein the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose of from about 20mg to about 200mg (e.g., 20mg, 60mg, 100mg, 160mg, or 200 mg) once every three weeks (e.g., on day 1 of a 21 day cycle). In certain embodiments, the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose of from about 10mg to about 100mg twice every three weeks (e.g., on days 1 and 8 of a 21 day cycle). In certain embodiments, the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose of from about 10mg to about 100mg once every two weeks (e.g., on days 1 and 15 of a 28 day cycle). In certain embodiments, the instructions further specify that one or more chemotherapeutic agents (e.g., carboplatin or cisplatin and/or etoposide) are administered to the subject in combination with the anti-DLL 3 and anti-PD-L1 agents. In various embodiments, the package insert may specify a maintenance period for administration of an anti-DLL 3 agent (e.g., AMG 757) in combination with chemotherapy, followed by administration of the anti-DLL 3 agent plus the anti-PD-L1 agent. Alternatively, the package insert may specify that an anti-DLL 3 agent (e.g., AMG 757) is administered in combination with an anti-PD-L1 agent as a maintenance-only therapy following standard-of-care chemoimmunotherapy. In embodiments in which the anti-DLL 3 agent and the anti-PD-L1 agent are used only as maintenance therapies, the package insert may further specify that subjects who receive at least 4 cycles (e.g., between 4 and 6 cycles) of platinum-based chemotherapy, etoposide and the anti-PD-L1 agent, and who have not undergone disease progression are eligible, and that subjects who are unable to obtain a first line of anti-PD-L1 agent and who receive platinum-based chemotherapy and etoposide for 4 to 6 cycles are eligible. Furthermore, for maintenance-only therapy, the package insert may additionally specify that treatment on cycle 1, day 1, be initiated within 8 weeks after the last chemotherapy cycle is initiated. The package insert may further specify that if radiation is completed at least 7 days prior to day 1 of cycle 1 in which the anti-DLL 3 agent is administered, prophylactic craniocerebral radiation therapy is allowed to proceed, and the subject does not need a steroid to manage Central Nervous System (CNS) symptoms.
In various embodiments, the package insert may further indicate that the subject is hospitalized and monitored for up to about 48 hours (e.g., about 24 hours, 12 hours, or 8 hours) after administration of the anti-DLL 3 agent (e.g., AMG 757) in cycle 1 and/or cycle 2. For example, the package insert may indicate that the subject is hospitalized and monitored for up to about 48 hours (e.g., about 24 hours, 12 hours, or 8 hours) after the first two or three doses of anti-DLL 3 agent in cycle 1. In various embodiments, the package insert may further indicate that one or more cytokines (e.g., IL-6, IL-8, IL-10, TNF- α, and IFN- γ) of the subject are measured or detected following administration of the anti-DLL 3 agent (e.g., AMG 757) in cycle 1, and the subject is hospitalized and monitored for up to about 48 hours (e.g., about 24 hours, 12 hours, or 8 hours) when any level of any cytokine is above a normal reference level. For example, the package insert may indicate that the subject's IL-10 is measured or detected after administration of an anti-DLL 3 agent (e.g., AMG 757) in cycle 1, and the subject is hospitalized and monitored for up to about 48 hours (e.g., about 24 hours, 12 hours, or 8 hours) when the level of IL-10 is above a normal reference level. Cytokines can be measured or detected using methods known in the art.
In certain embodiments, an article of manufacture comprises: (a) a container containing an anti-DLL 3 agent (e.g., AMG 757); and (b) a package insert carrying instructions for treating a DLL3 positive cancer (or treating SCLC) in a subject by administering an anti-DLL 3 agent (e.g., AMG 757) in combination with an alkylating agent (e.g., lubidine), wherein the instructions specify that the anti-DLL 3 agent is to be administered to the subject at a dose from about 20mg to about 200mg (e.g., 20mg, 60mg, 100mg, 160mg, or 200 mg) once every three weeks (e.g., on day 1 of a 21 day cycle).
In various embodiments, the containers contain the anti-DLL 3 agent (e.g., AMG 757) in an amount of about 1mg, 5mg, 10mg, or 25mg, e.g., the anti-DLL 3 agent is provided as a sterile, single-use, preservative-free, lyophilized drug product, with each container (e.g., vial) containing 1mg, 5mg, 10mg, or 25mg of the anti-DLL 3 agent. In various embodiments, the instructions specify reconstituting the lyophilized drug with water for infusion. In various embodiments, the instructions specify that the subject is a human (e.g., a human with SCLC).
8. A subject
In various cases of the presently disclosed methods, the subject is a human subject. In an exemplary embodiment, the human subject has DLL3 positive cancer. In an exemplary embodiment, the DLL3 positive cancer is Small Cell Lung Cancer (SCLC) or neuroendocrine prostate cancer (NEPC). In an exemplary case, the human subject has SCLC, optionally with histologically or cytologically confirmed SCLC. In various aspects, the human is a male or female with SCLC and/or is greater than or equal to 18 years old. In an exemplary aspect, the human subject has been treated with platinum-based chemotherapy. In an exemplary aspect, the human subject has RR SCLC, optionally with progression or recurrence after at least one platinum-based chemotherapy with or without a PD-L1 inhibitor. In an exemplary aspect, the human subject has ES-SCLC, optionally histologically or cytologically confirmed ES SCLC. In an exemplary aspect, the human subject has ES-SCLC and has not received prior systemic treatment for ES-SCLC. In an illustrative example, a human subject has an eastern tumor cooperative group (ECOG) physical stamina of 0-1 (OKen et al, am J Clin Oncol [ journal of clinical oncology ]5:649-655 (1982.) in various aspects, the human subject has one or more brain metastases that have been treated. Human subjects suffer NEPC, such as nepc occurring in new metastases or treatments, optionally, subject is subjected to at least 1 line of prior systemic treatment, including platinum-containing regimens for new NEPC (if they did not receive prior diagnosis or treatment for prostate cancer at NEPC) or androgen signaling inhibitors (e.g., abiraterone, enzalutamide, darostaamine, and/or apamide) (if occurring in treatment; prior diagnosis of prostate cancer has been preceded by NEPC diagnosis).
9. Cancer of the human body
In various aspects, the cancer treated by the presently disclosed methods is a DLL3 positive cancer (e.g., SCLC and NEPC). In various cases, the cancer treated by the methods disclosed herein is lung cancer, such as Small Cell Lung Cancer (SCLC). In an exemplary aspect, SCLC is histologically or cytologically confirmed SCLC. Alternatively, SCLC may be measured by revising the solid tumor remission standard (RECIST) 1.1, wherein the measurable lesions include (a) non-nodular lesions with sharp boundaries that can be measured accurately and continuously in one dimension of the axial plane (longest diameter. Gtoreq.10 mm, measured by magnetic resonance imaging/computed tomography (MRI/CT), scan slice thickness. Ltoreq.revising 5 mm) and/or (b) nodular lesions with longest diameter. Gtoreq.15 mm perpendicular to the long axis (short axis) according to MRI/CT, and/or exclude simple cysts, pleural/pericardial effusions and ascites. In various embodiments, the cancer treated by the presently disclosed methods is neuroendocrine prostate cancer (NEPC), such as NEPC that occurs in new metastases or treatments. In an exemplary embodiment NEPC is a histologically diagnosed minicell NEPC, or a prostate cancer with neuroendocrine differentiation, as defined by: positive immunohistochemical staining of chromogranin and/or synaptocins or ≡2 changes in Tp53, RB1 and PTEN in most tumor samples (by Immunohistochemistry (IHC), or genomic analysis of baseline tumor tissue or circulating tumor DNA (ctDNA)).
Examples
Example 1 clinical experience Using AMG 757
Study 20160232 is an open label, ascending, multi-dose, phase 1 study that evaluates AMG 757 in subjects with SCLC. There are two indications for this study: a: recurrent/refractory small cell lung cancer (RR SCLC) and B: extensive disease SCLC (ED SCLC).
The main end point is: dose Limiting Toxicity (DLT), adverse Events (AEs) occurring in treatment, treatment-related AEs, and clinically significant changes in vital signs, ECG, physical examination, and clinical laboratory testing.
Secondary endpoint: for indications a and B: (1) PK parameters of AMG 757 after intravenous administration, including, but not limited to, maximum observed concentration (C max), minimum observed concentration (C min), area under the concentration-time curve (AUC) over a 2 week dosing interval, accumulation after multiple dosing, and, if applicable, half-life (t 1/2), (2) Objective Remission (OR) according to the revised solid tumor remission assessment criteria (RECIST) 1.1, (3) remission Duration (DOR), and (4) 1 year Progression Free Survival (PFS) and (5) 1 year total survival (OS). Only for indication B: relapse Free Survival (RFS).
Exploratory endpoint: for indications a and B: (1) The incidence of anti-AMG 757 antibody formation, (2) changes in proteins, nucleic acids, and cellular biomarkers (e.g., cytokines, lymphocyte status, CTCs, sDLL 3) in the blood; (3) Cell surface protein expression (e.g., DLL 3) and tumor infiltrating lymphocyte status in tumor tissue at baseline. Only for indication B: effect of prior chemotherapy on AMG 757 pre-treatment T-cytokine production. CRS incidence (only for part of evaluating CRS mitigation strategies).
The key qualification criteria for study 20160323 are summarized in table 3 below.
TABLE 3 Critical qualification criteria
AMG 757 (0.003 mg to 100.0 mg) was administered intravenously in every two weeks.+ -. Step dosing in SCLC patients who progressed after receiving ≡1 platinum-based regimen. Antitumor activity was assessed using revision 1.1. The Progression Free Survival (PFS) and total survival (OS) were estimated using the Kaplan-Meier method. Tumor DLL3 expression was assessed by immunohistochemistry. T cell activation and cytokine profile were evaluated. AMG 757 or talastatin administration continues until disease progression occurs, unacceptable side effects occur, or informed consent is withdrawn.
Patients included in the ascending and expanding cohorts were analyzed. The date of expiration was 2022, 7, 19. A two-parameter Bayesian Logistic Regression Model (BLRM) model directs dose exploration. Security data is continuously reviewed. In a dose level review conference (DLRM), sponsors negotiate with on-site researchers to review BLRM recommended dose levels and all available cumulative data in a cohort prior to making dose escalation decisions. AE and DLT observed in all subjects were continuously assessed and fully incorporated into all DLRM. Based on the overall benefit-risk profile of 100mg, it was decided to evaluate it further as an extended dose. Descriptive statistics are provided for selected demographic, safety, pharmacokinetic (PK), pharmacodynamic and biomarker data. The median and percentile of the time to event endpoint was estimated using Kaplan-Meier, where Confidence Intervals (CIs) were calculated using Brookmeyer and Crowley methods.
The Maximum Tolerated Dose (MTD) is the highest dose considered safe that researchers and research teams jointly determine taking into account the Bayesian Logistic Regression Model (BLRM).
Adverse events were ranked using adverse event common terminology standard (CTCAE) version 4.0. CRS events were ranked using Lee criteria.
In addition, cytokine Release Syndrome (CRS), neutropenia, and nervous system events were monitored as events of interest in this study using AMGEN MEDDRA Query Narrow (AMQN) search methods. All events were encoded using MedDRA version 24.1. The cytokine release syndrome searched for AMQN includes cytokine abnormality, cytokine release syndrome, cytokine storm and cytokine detection. CRS events were ranked using the criteria of CRS Lee et al (2014). Neutropenia was searched based on AMQN and graded using CTCAE version 4.0. Nervous system events were searched based on "central neuropsychiatric event caused by direct neurotoxicity" AMQN and ranked using CTCAE version 4.0.
Efficacy data disclosed herein is based on local investigator assessment. A patient is defined as efficacy assessable if the data expiration date is at least 9 weeks after the first dosing date for time to assess.
Exploratory analysis of T cells and peripheral cytokines was performed on continuously collected blood samples.
For immunogenicity assessment, blood samples from patients receiving talastatin were collected at study day 1 (pre-dosing) and at various time points during the study to detect anti-talastatin binding antibodies using a validated electrochemiluminescent bridging immunoassay.
Immune cell and cytokine analysis
Whole blood samples drawn into EDTA tubes were collected according to the assessment plan specified in the study protocol. Whole blood samples were stained with fluorescence labeled antibodies CD4 BV510 (clone SK3, BD Biosciences), CD8 BV605 (clone SK1, BD Biosciences), CD3 Alexa Fluor 700 (clone SK7, bioleged) and CD279 (PD-1) BB515 (clone EH12.1, BD Biosciences) using a flow cytometry validated combination. Data was collected centrally on a BD FACSCanto flow cytometer by Q2 Solutions Laboratories Europe. To assess cytokine production, serum samples were collected and ifnγ levels were assessed using Meso Scale Discovery (MSD) V-plex pro-inflammatory combination 1. The assay was performed according to the manufacturer's instructions. Briefly, the samples were diluted with diluent 2 (MSD) in a 1:2 ratio. Diluted samples and standards were added in duplicate to 96-well plates with capture antibodies individually pre-coated on 10 defined spots and incubated for 2 hours at ambient temperature. Plates were washed three times with wash buffer and detection antibody mixtures were added to each well and plates were incubated for 2 hours at ambient temperature. Plates were washed three times with wash buffer and 2x read buffer T (MSD) was added to each well and read on an MSD microplate reader. The concentration was estimated to be within a predetermined range of 2.61-542,720pg/mL from the standard curve.
Characterization of Cytokine Release Syndrome (CRS)
Analysis was performed to explore the correlation between cytokine levels within 24 hours after the initial dose of talastatin and CRS occurring in cycle 1. Included in this analysis are a cohort receiving 1mg of talastatin as an initial dose in cycle 1 and a cohort receiving subsequent doses of 1mg to 100 mg.
Serum was withdrawn at time points up to 24 hours for cytokine analysis. CRS was evaluated for incidence, time of onset, severity, management, and recurrence. In cycle 1, peak serum levels and rate of rise within 24 hours after the initial dose of talastatin were evaluated for a set of soluble factors for patients with CRS versus patients without CRS. Including patients from a 1mg initial administration cohort.
A Kruskal Wallis (KW) rank test based on error-finding rate correction was used to identify whether analyte values from CRS and CRS-free categories originated from different distributions. The increasing trend of analyte values from CRS free to CRS group was determined using the Jonckheere-Terpstra (JT) trend test and error-finding rate correction. Single variable logistic regression was used to determine whether the rate of increase and peak capacity of each analyte predicted CRS to occur in cycle 1 after dosing.
Results
By day 19 of 7 months of 2022, 107 patients received talastatin in up-dosing (0.003 mg to 100mg; n=73) and expansion (100 mg; n=34) cohorts (fig. 1). Since Cytokine Release Syndrome (CRS) was observed in the previous cohort, step dosing was used starting from the 3mg cohort (1 mg was used as the infusion dose, followed by the target dose on day 8, day 15, and thereafter Q2W). Cytokine Release Syndrome (CRS) is an expected risk of talastatin according to its mechanism of action (MOA).
The baseline characteristics are summarized in table 3. The median age was 63 years (range 32 to 80 years). ECOG physical status was 0 to 1 in 99% of patients. More than 70% of patients received ≡2 line prior therapy, 25% of patients were refractory to platinum, and 50% of patients received prior PD-1/PD-L1 inhibitors.
The median follow-up period was 8.7 months (ranging from 0.2 to 31.8 months). 92 patients (86%) stopped treatment, most commonly due to disease progression (n=77 [72% ]). At the time of data cutoff, 47 patients (43.9%) ended the study with death. The median treatment period started was 3 (quartile range [ IQR ]:1, 8), and the median received Taraxazumab dose was 6 (IQR: 3, 16).
Table 3 patient demographics and baseline characteristics
* The disease phase at baseline was unknown for 1 patient.
ECOG, eastern tumor collaboration group in the united states; IQR, quartile range; PD-1, programmed cell death protein 1; PD-L1, programmed death ligand 1.
Retrospective DLL3 immunohistochemical analysis was performed on fresh or archived biopsy samples as described above. DLL3 was expressed in 85 out of 90 (94%) evaluable patients (. Gtoreq.1%); median H score was 186 (range 0 to 300) and median tumor cell positive rate was 95% (range 0% to 100%).
Safety and tolerability
DLT occurred in 6 patients, including pulmonary inflammation (n=1 [ last prior dose, 0.3mg ]), elevated alanine aminotransferase (n=1 [1mg ]), CRS (n=1 [1mg ]), encephalopathy (n=1 [10mg ]), chills, fever and neutropenia (n=1 each [100mg ]).) did not reach Maximum Tolerated Dose (MTD), the highest dose (100 mg) was evaluated in the expansion cohort (3.7%) in 4 patients, as a result of increased brain disease (n=1), immune effector cell-related neurotoxicity (ICANS) (n=1) and pulmonary inflammation (n=2), and the like, all of which were recorded as single G5 pulmonary inflammatory events in a treatment-related, in a 70 year old male with prior carboplatin/etoposide chemotherapy, chronic obstructive pulmonary disease, and pulmonary nodule and pleural nodule radiotherapy history on day 18 of cycle 1, a second single anti-therapeutic dose (3.7%) in the expansion cohort, and as a result of acute pulmonary inflammation (3.2) in the end of clinical progression of both clinical conditions (3 mg) in the three patients, as a result of increased pulmonary inflammation (3.7%) in the other cases of acute pulmonary inflammation and pulmonary inflammation (pulmonary inflammation) in the case of clinical conditions, as well as a clinical progression of pulmonary inflammation (pulmonary inflammation) in the three cases of the acute pulmonary inflammation (pulmonary inflammation) in the case of the three patients and the pulmonary inflammation (pulmonary inflammation) in the case of the clinical condition (pulmonary inflammation) and the clinical condition (pulmonary inflammation) was observed in the clinical cases of the clinical conditions of the four cases. 1 patient regressed lung inflammation before withdrawal from PD, and 1 patient resumed treatment with unchanged dose.
Any cause/grade TEAE occurred in 107 patients (100%). The most common are CRS (56 patients [52.3% ]), fever (43 cases [40.2% ]), constipation (33 cases [30.8% ]) and fatigue (32 cases [29.9% ]). 61 patients (57.0%) developed grade 3 AE, most commonly neutropenia (8.4%), lymphocyte count reduction (6.5%) and hypertension (5.6%). Serious Adverse Events (SAE) occurred in 55 patients (51.4%). TEAE resulted in a dose reduction in 9 patients (8.4%), with CRS-related reduction occurring in 4 (3.7%). Dose interruption occurred in 20 patients (18.7%), most common causes were neutropenia and a decrease in neutrophil count. 97 (90.7%) and 33 (30.8%) patients developed any grade and ≡3 grade TRAE, respectively.
CRS, neutropenia, and neurological events were monitored as events of interest based on preclinical, clinical, and mechanistic data using talastatin, other BiTE TM molecules, and other T cell-related therapies. A AMGEN MEDDRA Query Narrow (AMQN) search is performed to supplement the standard system organ category single preferred term security report (as defined above and summarized in table 4). Measures to mitigate the likelihood of CRS include prophylactic corticosteroids (cycle 1 only) and IV supplementation in some patients. 15 patients (14.0%) reported CRS occurring in ≡2 treatment, and 1 patient (0.9%) reported CRS of grade 3; there are no class 4 or class 5 CRS reports. For any fractionated CRS (n=56), the median time to first episode was 2 days (ranging from 1 day to 30 days) after the first dose based on the date recorded; a more accurate time-based report was performed to better characterize CRS, with median seizure time of 17.5 hours in the subset of patients with available hourly data (n=47). CRS was transient (median duration 3 days [ IQR:2 days to 4 days ]), and regressed in all cases. 8 patients (7.5%) received tolizumab for CRS. CRS is mainly limited to cycle 1. A total of 5 patients (4.7%) developed CRS during cycle 2; of these, 4 patients also developed CRS in cycle 1, while 1 patient experienced CRS for the first time in cycle 2 or later. 75 patients (70.1%) developed any grade of neurological AE that occurred in treatment, and most of the neurological AE were grade 1; the most common are dysgeusia (29.0%), headache (19.6%) and dizziness (10.3%). 12 patients (11.2%) had developed neurological events that occurred in grade 3 treatment, including confusion (4.7%), delirium (1.9%) and encephalopathy (1.9%). class 4 neurological events (confusion) occurred in 1 subject, and class 5 neurological events occurred in no patient. All ≡3 grade neurological AEs resolved, with 1 subject disabling talastatin due to G3 encephalopathy and the other 2 subjects continuing treatment with reduced doses. G2 ICANS is another neurological cause that results in withdrawal in 1 subject. The first onset of any graded neurological event occurred mostly within the first 30 days of treatment (median 9 days [ IQR,2 days to 29 days ], with median duration of 5 days (IQR, 2 days to 15 days). 11 patients (10.3%) had ≡3 grade neutropenia. The median time to onset of any graded neutropenia first occurred 30 days (IQR, 21 days to 31 days) after the first time of talastatin administration, and the median duration was 7 days (IQR, 4 to 13); overall, 10 patients (9.3%) received G-CSF. Febrile neutropenia occurred in 1 patient, but it was considered irrelevant to treatment.
TABLE 4 adverse events occurring in treatment (AMQ of preferred and selected terms)
* Based on the occurrence of a single preferred term. Version meddra25.0 was used. Adverse event grading was performed using CTCAE version 4.0 and CRS event grading was performed using the standard of Lee et al (2014).
CRS are based on AMQ narrow searches including cytokine abnormalities, cytokine release syndrome, cytokine storms, cytokine detection. Neutropenia is based on AMQ narrow-range searches. Nervous system events are based on an "immediate neurotoxic induced central neuropsychiatric event" AMQ narrow search. Version meddra25.0 was used. CRS event ranking was performed using the criteria of CRS Lee et al (2014). Neutropenia and neurological event classification was performed using CTCAE version 4.0.
AMQ, amgen MedDRA queries.
Efficacy of
The ORR was confirmed to be 23.4% (95% confidence interval [ CI ]:15.7, 32.5), including 2 complete remissions and 23 partial remissions (Table 5).
TABLE 5 tumor remission by Taraxazumab according to the investigator's assessment
* The interim efficacy analysis set is a subset of the safety analysis set. The set of metaphase efficacy analyses includes patients with a data cutoff date of at least 9 weeks after the date of first administration.
This includes 32 patients who developed PD in the post-baseline scan but did not further confirm the scan (unidentified PD according to revision 1.1).
Reasons for not performing imaging evaluations include withdrawal of informed consent (n=5), death (n=2), clinical PD (n=1), and initiation of new anti-cancer therapies (n=1).
Fig. 1A illustrates the optimal percent change from baseline in the sum of diameters of patients (n=94) with an assessable post-baseline assessment result. The disease control rate was 51.4% (95% CI:41.5, 61.2). Remission is seen starting from the 0.3mg dose, and higher remission rates are generally observed at 3mg and higher doses. Tumor shrinkage in target lesions was observed in 39 patients (36.4%) at least 30% upon post-baseline assessment. In the confirmed remittes, median TTR was 1.8 months (range 1.2 to 7.4), and median DOR was 12.3 months (95% CI:6.6, 14.9) (FIG. 1B). The maximum duration of remission was 14.9 months, and 11 patients (44% of remittes) had ongoing remissions at the time of data cutoff. Median PFS was 3.7 months (95% CI:2.1,5.4) and median OS was 13.2 months (95% CI:10.5, NE), respectively (FIG. 2). A total of 28 patients (26.2%) continued to receive subsequent anti-cancer therapy after talastatin.
Clinical pharmacokinetics
By day 15 of 4 of 2022, preliminary pharmacokinetic data for 101 patients were obtained from the up-dosing and expansion cohorts. In short, talastatin appears to increase approximately proportionally to the dose in terms of serum exposure. Within 4 weeks after the start of the weekly target regimen, the serum taratamab exposure reached near steady state with minimal accumulation. The estimated average (+ -SD) terminal elimination half-life at steady state is about 5.7 (+ -2.2) days within the estimated target dose range, which is consistent with the expected prolonged half-life of HLE platform relative to non-HLE BiTE TM molecules.
Immunogenicity of
Of the patients with available samples, 10 out of 97 patients (10.3%) produced anti-taraxamab antibodies after administration of taraxamab. Two of the 99 patients (2.0%) had pre-existing antibodies at baseline. In these patients, the anti-drug antibody (ADA) had no significant effect on the amount of taratamab exposure or on the safety profile.
Pharmacodynamics of medicine
The pharmacodynamic response following infusion of the first dose of talastatin is characterized by initial T cell redistribution, T cell activation and transient IFN- γ elevation. For the step dose cohort, the pharmacodynamic response was maximal after initial administration of the 1mg step dose, and the results were not exceeded when administered at the target dose.
Summary of clinical CRS
CRS was mostly grade 1 (39%), occurred at cycle 1, and was reversible in all patients (see table 6). CRS is clinically manageable.
TABLE 6
To mitigate risk of CRS, the following preventive measures can be used during cycle 1, which is ≡1:: by using
Oral dexamethasone for additional corticosteroid prophylaxis, administration of Tozumazumab, etanercept or pair
Acetaminophen.
* CRS includes cytokine abnormality, cytokine release syndrome, cytokine storm, and cytokine
And (5) sub-detection.
Percentages are based on the total number of patients with any fractionated CRS.
The time of onset of the patient (n=47) with both date and time of onset data.
Data cut-off: 2022, 6 and 15.
CRS, cytokine release syndrome.
Cytokine and CRS analysis
Among patients with evaluable biomarkers, the ratio of peak to baseline levels in 24 hours for IL-6, IL-8, IL-10 and TNF-a tended to be higher in patients with CRS in cycle 1 than in patients without CRS (fig. 3A-3D). IL-10 showed a significantly higher elevation than the reference normal range and was higher in patients with CRS (fig. 4). Because of its strong induction in preclinical trials, the effector cytokine IFN-gamma was studied (see, e.g., giffin MJ et al CLIN CANCER RES [ clinical cancer research ]2021; 27:1526-1537). As expected from the mechanism of action of talastatin, IFN- γ induction is above the physiological range; similar induction was induced between patients with period 1 CRS and patients not with period 1 CRS (fig. 5A and 5B).
Discussion of the invention
With an extended dose of 100mg, taratamab exhibits manageable safety profiles over a broad dose range and is associated with encouraging remission rates in a largely pretreated SCLC patient population. The mitigation of the validation is persistent and appears to be promising to implement the OS. Of all doses (n=107), only 4 patients (3.7%) were discontinued from talatizumab, and 9 patients reduced the dose due to AE. The MTD is not reached; the highest dose (100 mg) was further evaluated in the dose expansion cohort.
CRS is expected to occur based on MOA of talastatin. While CRS is the most common TEAE observed in this study (56% of patients), it is typically low-grade, transient, and usually occurs in the first cycle. CRS is generally reversible and managed by steroid, IV infusion and antipyretics, with 8 out of 107 patients receiving talastatin (7.5%) treated with tolizumab. Neutropenia is a risk associated with talastatin observed in this study and is unexpected based on preclinical data; the mechanism is unknown. Thus, research schemes have been updated for specific monitoring and management. Further evaluation of neutropenia will be relevant to experiments using talastatin in combination with other myelosuppressive therapies. Since it is known to be associated with immune effector cell therapy, neurological assessment is performed as part of frequent clinical assessment to assess CRS and/or neurological AE of study patients. Although 12 patients (11.2%) developed ≡3 grade neurological AE, most neurological AE were mild and self-limiting without stopping treatment or reducing dose. There were 2 patients who had been refractory to the neurological AE (encephalopathy, ICANS) to talastatin. Careful assessment of the nervous system AE is underway to better characterize these events and to determine risk factors or interventions that can specifically improve management.
There are few approved post-line SCLC therapies. Phase 2 studies on lubidine in two-line SCLC found an ORR of 35% and a median DOR of 5.3 months. In a randomized study of topotecan in recurrent SCLC with combination chemotherapy, the ORR of topotecan was 24% and the median DOR was 3.3 months. Previous conditional approval of nivolumab and pembrolizumab by the U.S. FDA for tri-line or later SCLC is based on remission rates of 12% and 19%, respectively, with sustained remissions of > 12 months observed in >60% remission patients. These approvals are then withdrawn as no survival benefits are demonstrated. The DOR of talastatin was 23%, the median DOR was 12.3 months, good compared to other therapies, especially considering that more than 70% of patients received at least 2 line-preceding normals. Half of the patients in this study (50%) received prior PD-1/PD-L1 therapy, which represents current practice for first-line SCLC. Although median PFS (3.7 months) was observed with talastatin, median OS (13.2 months) was relatively high and superior to previously reported median OS with lubidine (9.3 months) or OS with topotecan (about 6 months), the value of the comparison was limited by study design and patient population differences. The promising OS benefit may reflect the long-term persistence of relief observed so far in those patients who respond to talastatin, but need to be further followed in larger scale random studies. An alternative explanation for the relatively longer OS and shorter PFS may be that OS benefit from post-talastatin treatment, but this is unlikely to be the major factor, since only 26.2% of patients in this largely pre-treated cohort received such treatment. Efforts are underway to determine clinical, demographic, and biological factors (e.g., prior therapies, DLL3 expression) that can predict response and/or toxicity. Increased DLL3 expression appears to tend to have a higher degree of clinical benefit.
The results of this example demonstrate the promising activity of talastatin in patients with highly unmet medical needs and led to several ongoing studies of talastatin as monotherapy for SCLC and other neuroendocrine cancers.
Example 2 prolonged intravenous infusion study design of AMG 757 in phase 1 study assessing safety, tolerability, and pharmacokinetics of AMG 757 in subjects with small cell lung cancer
Study 20160232 is an open label, ascending, multi-dose, phase 1 study that evaluates AMG 757 in subjects with SCLC. There are two indications for this study: a: recurrent/refractory small cell lung cancer (RR SCLC) and B: extensive disease SCLC (ED SCLC).
The primary, secondary, exploratory endpoints and key inclusion and exclusion criteria for this study are listed in example 1.
To reduce the incidence of CRS, cycle 1 dosing schedules were adjusted as described herein to administer AMG 757 over a longer infusion duration. Specifically, AMG 757 is administered for cycle 1 day 1 dosing by prolonged intravenous (eIV) infusion (e.g., infusion duration ranging from 2 days to 7 days) to reduce the incidence and/or severity of CRS. eIV infusion administration involves infusion of an introduced dose or target dose administered via 2, 3,5, or 7 day eIV infusion, followed by a step dose (equal to the target dose) bolus IV infusion on day 8 (e.g., a 60 minute IV infusion) and a target dose bolus IV infusion on day 15, or followed by a target dose bolus IV infusion on day 15. Dose levels for the eIV method beginning on day 1 of cycle 1 include infusion of a dose from about 1mg to about 200mg (e.g., 30mg or 100 mg) over a 72 hour duration followed by bolus IV infusion on days 8 and 15 or only on day 15. Thereafter, AMG 757 is administered at Q2W (e.g., beginning on day 29), twice every three weeks (e.g., beginning on day 22), or at Q3W (e.g., beginning on day 22). A summary of eIV dosing schedules (cycle 1 only) is shown in the table below.
EXAMPLE 3 prolonged IV administration to patients with SCLC and results
SCLC patients were placed into a group eIV cohort for AMG 757 monotherapy dose exploration and expansion. Thirty-one (31) patients were placed in group eIV cohorts by day 1,3, 2023. Starting on day 1 of cycle 1, 30mg or 100mg of AMG 757 was administered via eIV infusion over a period of 3 days (72 hours), followed by bolus infusion of AMG 757 at doses of 100mg on days 8 and 15 of cycle 1, followed by administration at 100mg once every two weeks. Of 31 patients, 6 received 30mg eIV administrations (cohort 26) and 25 received 100mg eIV administrations (cohorts 27 and 31). On cycle 1, day 1, dexamethasone 8mg IV (or equivalent) was administered 1 hour prior to the start of AMG 757 infusion. In addition, saline (1L) was administered for 4 to 5 hours from the beginning of the infusion of AMG 757 on day 1 of cycle 1. The treatment results are summarized in table 7 below.
TABLE 7 summary of objective remission and tumor shrinkage for eIV administration
CI = confidence interval; n = number of subjects in the mid-term efficacy analysis set; KM = kaplan-meyer; n1=the number of confirmed mitigators; n2=disease control population (cr+pr+sd).
The interim efficacy analysis set is a subset of the safety analysis set. The interim efficacy analysis set includes subjects with a data cutoff date of at least 9 weeks after the first dosing date.
a The assessment of disease response is determined according to the revision 1.1 guidelines.
b "Unconfirmed PD" classifies subjects who reached PD in a post-baseline scan but who did not undergo further confirmation of the scan.
c The exact 95% confidence interval was calculated using the Clopper Pearson method.
d Including subjects with any tumor shrinkage in the target lesion at the time of post-baseline assessment.
e Including subjects with at least 30% tumor shrinkage in the target lesion at the time of post-baseline assessment.
f The minimum and maximum values of the mitigation duration use only event observations and do not include deletion observations.
Snapshot date: 2023 1 month 3. Date of data expiration: 2023 1 month 3.
Adverse events occurring in any of the graded treatments were observed in all patients in the eIV cohort. Adverse events resulted in 1 patient (3.8%) of AMG 757 dose reduction and 1 patient (3.8%) withdrawal in cohorts 27 and 31. They do not result in dose reduction or withdrawal for any of the patients in the cohort 26.
Example 4 evaluation of safety, tolerability and pharmacokinetics of AMG 757 in phase 1 study of AMG 757 in subjects with small cell lung cancer three week dosing regimen
Study 20160232 is an open label, ascending, multi-dose, phase 1 study that evaluates AMG 757 in subjects with SCLC. There are two indications for this study: a: recurrent/refractory small cell lung cancer (RR SCLC) and B: extensive disease SCLC (ED SCLC).
The primary, secondary, exploratory endpoints and key inclusion and exclusion criteria for this study are listed in example 1.
Two 21-day dosing regimens of AMG 757 were studied in subjects with SCLC. Beginning at cycle 2, the subject receives AMG 757 twice every three weeks for a 21 day cycle (e.g., on days 1 and 8 of the 21 day cycle) or once every three weeks for a 21 day cycle (e.g., on day 1 of the 21 day cycle). Step doses are administered in cycle 1, such as a first step dose on day 1, a step dose equal to the target dose on day 8, and a target dose on day 15.
Day 1 and day 8 (D1/D8) of the 21 day cycle: subjects received 1mg of AMG 757 on day 1 followed by 100mg of target doses on days 8 and 15. From cycle 2, day 1, the subject received the target dose of AMG 757 on days 1 and 8. The initial target dose of AMG 757 was 100mg.
Dosing once every 3 weeks (Q3W): subjects received 1mg of AMG 757 on cycle 1, day 1, followed by a step dose on day 8, and a target dose on day 15. From and after cycle 2, day 1, the subject received the target dose at Q3W. The initial target dose of Q3W was 200mg. Q3W dosing schedules can provide improved convenience and flexibility for patients and healthcare workers. It also has a positive impact on resource utilization (e.g., fewer days in a month occupied by the treatment center). Subjects in the 200mg q3w cohort received 100mg AMG 757 on cycle 1, day 8. The dose-decreasing target doses administered by AMG 757Q3W included 100mg Q3W and 60mg Q3W.
By day 1,3 of 2023, 11 patients received treatment in the Q3W dosing cohort (cohort 37), and 1 patient received treatment in the day 1 and day 8 cohorts (cohort 38) of the 21 day cycle. Briefly, for Q3W administration, 1mg of AMG 757 was administered on day 1 of cycle 1, then 100mg was administered on day 8 of cycle 1, and 200mg was administered on day 15 of cycle 1. 200mg of AMG 757 was administered at Q3W beginning on and after day 1 of cycle 2. For D1/D8 administration, 1mg of AMG 757 was administered on day 1 of cycle 1, followed by 100mg on days 8 and 15 of cycle 1. 100mg of AMG 757 was administered at D1/D8 for the 21 day period beginning on and after day 1 of the 2 nd period. Patients receiving D1/D8 dosing respond as stable disease. The results of treatment for patients receiving Q3W administration are summarized in table 8 below.
TABLE 8 summary of objective remission and tumor shrinkage for AMG 757Q3W administration
CI = confidence interval; n = number of subjects in the mid-term efficacy analysis set; KM = kaplan-meyer; n1=the number of confirmed mitigators; n2=disease control population (cr+pr+sd).
The interim efficacy analysis set is a subset of the safety analysis set. The interim efficacy analysis set includes subjects with a data cutoff date of at least 9 weeks after the first dosing date.
a The assessment of disease response is determined according to the revision 1.1 guidelines.
b "Unconfirmed PD" classifies subjects who reached PD in a post-baseline scan but who did not undergo further confirmation of the scan.
c The exact 95% confidence interval was calculated using the Clopper Pearson method.
d Including subjects with any tumor shrinkage in the target lesion at the time of post-baseline assessment.
e Including subjects with at least 30% tumor shrinkage in the target lesion at the time of post-baseline assessment.
f The minimum and maximum values of the mitigation duration use only event observations and do not include deletion observations.
Snapshot date: 2023 1 month 3. Date of data expiration: 2023 1 month 3.
Adverse events occurring in any of the graded treatments were observed in 10 patients (83.3%) in cohort 37, with 1 patient having a reduced AMG 757 dose and 1 patient discontinuing medication due to adverse events. Adverse events occurring in any of the graded treatments were observed in 3 patients (100%) in cohort 38, with no dose reduction or withdrawal.
Example 5 phase 1 study to evaluate the safety and efficacy of first line talastatin (AMG 757) in combination with carboplatin, etoposide and PD-L1 inhibitors in subjects with extensive stage small cell lung cancer
The present study (study 20200469) is a phase 1b, multicenter, open-label study, assessing safety, tolerability, PK, pharmacodynamics (PD) and primary efficacy of talatient in combination with standard-of-care chemo-immunotherapy in subjects with ES-SCLC. Taraxamab was evaluated as follows: a maintenance cycle of talastatin plus anti-PD-L1 (e.g., alemtuzumab) followed by induction of chemotherapy plus anti-PD-L1 (e.g., alemtuzumab), and as a maintenance-only therapy, wherein talatamab is administered in combination with anti-PD-L1 (e.g., alemtuzumab) after standard-of-care chemotherapy. The indication for this study was SCLC.
The targets and endpoints of this study are listed in table 9 below.
TABLE 9
The key inclusion and exclusion criteria are summarized below.
Critical inclusion criteria
Age equal to or greater than 18 years old
Subjects suffering from histologically or cytologically confirmed ES SCLC and not receiving prior systemic treatment for ES-SCLC except for the first line therapy described below. Subjects who are allowed to receive prior treatment for localized SCLC
Part 1 to part 4 and part 7: the subject must have received 1 cycle of platinum chemotherapy, etoposide and PD-L1 inhibitor. Subjects who did not obtain PD-L1 inhibitors were eligible.
Part 5, part 6, part 8 and part 9: the subject must have received between 4 and 6 cycles of first line platinum chemotherapy, etoposide and PD-L1 inhibitor and no disease progression occurs. Subjects who received 4 to 6 cycles of platinum chemotherapy plus etoposide were eligible if no first line PD-L1 inhibitor was obtained.
Disease measurable by revision solid tumor Remission Evaluation Criteria (RECIST) 1.1 (except part 5, part 6, part 8, and part 9).
Eastern tumor collaboration group 0 to 1
Subjects with treated asymptomatic brain metastases are eligible as long as the criteria of the regimen are met
According to the adequate organ functions determined by the local laboratory, the following are defined:
Absolute count of neutrophils not less than 1.5x10 9/L
Platelet count of ≡100x10 9/L
Hemoglobin of 9g/dL or more
Estimated glomerular filtration rate >60mL/min/1.73m based on renal disease diet adjustment calculation 2
Aspartic acid aminotransferase and alanine aminotransferase are less than or equal to the upper limit of 3 Xnormal (ULN) (or less than or equal to 5 XULN for subjects with liver involvement)
Total bilirubin is less than or equal to 1.5 XULN (or less than or equal to 2 XULN for subjects with liver metastases)
Prothrombin Time (PT)/International normalized ratio and partial thromboplastin time or activated partial thromboplastin time.ltoreq.1.5 x institution ULN
Lung function:
there was no clinically significant pleural effusion on study day 1. Allowing for treatment of pleural effusion to meet the conditions.
Baseline oxygen saturation >90% under indoor air.
Heart function:
The heart ejection fraction is more than or equal to 50%
Critical exclusion criteria
Subjects were excluded from the study if any of the following criteria were met:
With untreated or symptomatic brain metastases and/or pia materia diseases
Medical history or evidence of interstitial lung disease or active non-infectious pneumonia
Is diagnosed as immunodeficiency or is receiving systemic steroid therapy or any other form of immunosuppressive therapy within 7 days prior to the first administration of the study treatment
Have a history of any immune-related colitis
The subject had symptoms and/or clinical signs and/or radiological signs indicative of acute and/or uncontrolled active systemic infection within 7 days prior to the first dose of study treatment. With history of pituitary inflammation or pituitary dysfunction
A history of arterial thrombosis (e.g., stroke or transient ischemic attacks) within 6 months of the group
Myocardial infarction and/or symptomatic congestive heart failure (New York Heart Association > class II in the United states) or unstable angina within 6 months of study day 1. Unstable cardiac arrhythmias were present within 3 months of study day 1. Has clinically significant pericardial effusion
Active autoimmune diseases that required systemic treatment (except replacement therapy) over 2 years or any other disease that required immunosuppressive therapy at the time the study was conducted. Subjects who are allowed to suffer from type I diabetes, vitiligo, psoriasis, hypothyroidism or hyperthyroidism without immunosuppressive treatment
Live vaccine therapy received 4 weeks prior to study drug administration
Have a history of solid organ transplants
Medical history or evidence of any other clinically significant disorder, condition or disease (other than those outlined above) that would appear to a researcher or an advanced (Amgen) physician (if consulted) to pose a risk to subject safety or interfere with research assessment, procedure or completion
Example 6 study design and results of study 2020469
Overall design: this is a phase 1, multicentric, open-label study that evaluates the safety, tolerability, PK, pharmacodynamics (PD), and primary efficacy of talastatin in combination with standard-of-care chemo-immunotherapy in subjects with ES-SCLC. Taraxamab was evaluated as follows: in combination with induction chemotherapy plus anti-PD-L1 (e.g., alemtuzumab), followed by a maintenance cycle of talatamab plus anti-PD-L1 (e.g., alemtuzumab), and as a maintenance-only therapy, wherein talatamab is administered in combination with anti-PD-L1 (e.g., alemtuzumab) after standard-of-care chemotherapy
Taraxamab was administered as a short-term Intravenous (IV) infusion over 60 minutes, followed by rinsing. To mitigate the risk of Cytokine Release Syndrome (CRS), a step dosing method was performed during the first cycle of administration of talastatin in each fraction. Three different dosing schedules for talatizumab were evaluated: once every 2 weeks (Q2W), on days 1 and 8 of the 21 day cycle (D1/D8), and once every 3 weeks (Q3W). For all dosing schedules, talastatin administration starts with 1mg administered on day 1 of cycle 1, followed by one or more stepped doses to achieve the target dose. The prodrug was administered with dexamethasone 8mg IV (or equivalent dose of other corticosteroid) 1 hour prior to all first periodic doses of talastatin. A prophylactic IV replacement fluid (e.g., 500 to 1000mL saline over about 4 to 5 hours) will also be administered after all of the taratamab cycle 1 doses.
The study consisted of 9 parts and included dose escalation (part 1, part 2, part 3 and part 5) and dose expansion (part 4, part 6, part 7, part 8 and part 9):
Part 1 (induction of Taraxamab D1/D8 and maintenance of Taraxamab Q2W): taraxazumab was combined with alemtuzumab, carboplatin and etoposide in cycle 1 starting with step dosing, followed by alemtuzumab D1/D8 in cycles 2 and 3 in combination with alemtuzumab, carboplatin and etoposide. From cycle 4, subjects received a maintenance cycle of atacamab q2w+ once every 4 weeks (Q4W) of talastatin.
The treatment regimen of part 1 is as follows:
Induction: cycle 1 (21 day cycle): on day 1, either 1200mg IV of alemtuzumab was administered followed by carboplatin to match the area under the concentration-time curve (AUC) 5IV, followed by 100mg/m 2 IV of etoposide. Taraxamab was administered after completion of chemotherapy. Taraxamab D1/D8 was started with a 1-step dosing, wherein 1mg IV of Taraxamab was administered on cycle 1, day 1. Etoposide 100mg/m 2 IV was administered on days 2 and 3. Taraxamab D1/D8 target doses were given on cycle 1, day 8 and day 15.
Cycle 2 and 3 (21 day cycle): on day 1, atractylizumab 1200mg IV was administered followed by carboplatin to match AUC 5IV and etoposide 100mg/m 2 IV. Taraxamab target doses were administered after completion of day 1 chemotherapy. Etoposide 100mg/m 2 IV was also administered on days 2 and 3. Taraxamab target doses were also administered on day 8.
Maintaining: period 4+ (28 day period): taraxamab Q2W target dose IV on day 1 and day 15. Ab bead mab 1680mg IV Q4W on day 1.
Part 1 includes combinations of 1 or more of the following dose levels of talastatin (table 10-1) with fixed doses of alemtuzumab administered according to standard of care, as well as carboplatin and etoposide.
TABLE 10-1 part 1 Taraxazumab administration
IV: intravenous; N/A: is not suitable for
A 21 day period
B 28 day period
Dose queue level 1.1 Taraxamab starts at 1mg at the first step dose on day 8 of cycle 1, followed by a target dose of 10mg on day 15 of cycle 1.
Part 2 (induction of talatimab Q3W and maintenance of talatimab Q3W): taraxazumab was combined with alemtuzumab, carboplatin and etoposide in cycle 1 starting with step dosing, followed by alemtuzumab Q3W in cycles 2 and 3 in combination with alemtuzumab, carboplatin and etoposide. From cycle 4, the subject received a maintenance cycle of talastatin q3w+atuzumab Q3W.
The treatment regimen of part 2 is as follows:
Induction: cycle 1 (21 day cycle): on day 1, atractylizumab 1200mg IV was administered followed by carboplatin to match AUC 5IV, and then etoposide 100mg/m 2 IV. Taraxamab was administered after completion of chemotherapy. On cycle 1, day 1, talab Q3W was started with a first step dose of talab (1 mg). Etoposide 100mg/m 2 IV was administered on cycle 1, day 2 and day 3. Taraxamab Q3W target doses were administered on cycle 1 day 8 and cycle 1 day 15 (see Table 10-2). Alternatively, the target dose of taratamab Q3W was administered on day 8 of cycle 1 and no treatment was performed on day 15 of cycle 1 (see table 10-2).
Cycle 2 and 3 (21 day cycle): on day 1, either 1200mg IV of atuzumab was administered followed by carboplatin to match AUC 5, and then 100mg/m 2 IV of etoposide. Taraxeumab Q3W target doses were administered after completion of day 1 chemotherapy. Etoposide 100mg/m 2 IV was also administered on days 2 and 3.
Maintaining: period 4+ (21 day period): the alemtuzumab 1200mg IV was administered on day 1 followed by the talastatin Q3W target dose on day 1 of the 21 day cycle.
TABLE 10-2 part 2 Taraxazumab administration
IV: intravenous; N/A: inapplicable; Q3W: every 3 weeks
Dose level 2.1 Taraxamab was administered beginning with a1 st step dose of 1mg administered on day 8 of cycle 1, a 20mg target dose administered on day 15 of cycle 1, and a 20mg Q3W target dose administered on day 1 of cycle 2.
Dose level 2.3 Taraxamab was administered starting with a1 st step dose of 1mg administered on day 1, a2 nd step dose of 100mg administered on day 8, and a target dose of 200mg administered on day 15.
If 200mg of Q3W is deemed unsafe, then 100mg of Q3W can be explored as an alternative dose level for Taraxazumab. Taraxamab was administered at 1mg on day 1, 100mg on day 8, 100mg on day 15, and 100mg on cycle 2, day 1.
In parts 1 and 2, depending on the security data observed, the following may occur: 1) dose decrementing to the next lowest dose cohort level, 2) additional cohorts to the current dose cohort level, or 3) dose escalating to the next highest dose cohort level or beginning dose expansion into the cohort. Allowing re-incrementing to the next higher dose queue level as appropriate. If a re-increment occurs, alternative (intermediate) dose cohort levels are explored as suggested by dose level review conference (DLRM), including adjusting the dose of talatimumab or adjusting the date of administration of talatimumab. Fraction 1 dose level 1.3 and fraction 2 dose level 2.3 were explored if a 100mg D1/D8 and 200mg q3w of talastatin monotherapy doses were considered safe.
Part 3 (induction of Taraxamab D1/D8 and maintenance of Taraxamab Q3W): taraxazumab was combined with alemtuzumab, carboplatin and etoposide in cycle 1 starting with step dosing, followed by alemtuzumab D1/D8 in cycles 2 and 3 in combination with alemtuzumab, carboplatin and etoposide. From cycle 4, the subject received a maintenance cycle of talastatin q3w+atuzumab Q3W.
The treatment regimen of part 3 is as follows:
Induction: cycle 1 (21 day cycle): on day 1, atractylizumab 1200mg IV was administered followed by carboplatin to match AUC 5IV, and then etoposide 100mg/m 2 IV. Taraxamab was administered after completion of chemotherapy. The highest safe and tolerogenic dose of Taraxamab D1/D8 was started on day 1 of cycle 1 with a 1mg step administration. Etoposide 100mg/m 2 was administered on days 2 and 3. Taraxamab was administered on cycle 1, day 8 and day 15.
Cycle 2 and 3 (21 day cycle): on day 1, atractylizumab 1200mg IV was administered followed by carboplatin to match AUC 5IV, and then etoposide 100mg/m 2 IV. Taraxamab D1/D8 target dose was administered after completion of day 1 chemotherapy. Etoposide 100mg/m 2 IV was also administered on days 2 and 3. Taraxamab target doses were also administered on day 8.
Maintaining: period 4+ (21 day period): the alemtuzumab 1200mg IV was administered on day 1 followed by the taletamab Q3W target dose on day 1 of each 21-day cycle.
TABLE 10-3 part 3 Taraxazumab administration
Part 4 (expansion of part 1, part 2 or part 3)
Part 5 (maintenance talatizumab Q2W plus alemtuzumab): taraxazumab was combined with alemtuzumab in cycle 1 starting with step dosing. From cycle 2, the subject received talastatin q2w+atuzumab Q4W.
Part 5 starts with a Taraxamab dose level of 5.1 (Table 10-4). If a combination of lower dose levels has been declared safe in part 1 or part 2, the group entry may be initiated at a dose level of 5.2 or 5.3.
The treatment regimen of part 5 is as follows:
Cycle 1 (28 day cycle): on day 1, alemtuzumab 1680mg IV was administered followed by talatamab. Taraxamab begins with a step dosing on cycle 1, wherein 1mg IV of Taraxamab is administered on cycle 1, day 1, followed by administration of Taraxamab Q2W target dose IV on days 8 and 15.
Period 2+ (28 day period): the Taraxamab target dose was administered on day 1 with 1680mg IV q4W followed by the Taraxamab target dose on days 1 and 15 from cycle 2.
TABLE 10-4 Taraxazumab administration
Part 6 (expansion of part 5)
Part 7: will be an extension of the combined dose using Dewaruzumab as a PD-L1 inhibitor to select an extension from part 1, part 2 or part 3. Dewaruzumab will be administered at a dose of 1500mg IV once every 4 weeks or at a dose of 1500mg once every 3 weeks.
Part 8: an extension of the combination dose cohort using Dewaruzumab as a PD-L1 inhibitor to select an extension from part 5. Dewaruzumab will be administered at a dose of 1500mg IV once every 4 weeks.
Part 9: an extended queue of talastatin-plus-valuzumab will be maintained, each administered once every 3 weeks. The dosage of Taraxamab Q3W will be selected from one of the dosage levels in Table 10-2. Dewaruzumab will be administered at a dose of 1500mg once every 3 weeks.
Dose escalation/decrementing recommendations were guided by a revised toxicity probability interval-2 (mTPI-2) model (Guo et al, 2017), with a target toxicity probability of 30%, an equivalent toxicity interval (25%, 35%), and an overdose probability of 95%. Beta (1, 1) is used as a priori distribution.
Results
By day 1 and 3 of 2023, a cohort of 15 patients (5 patients in cohort 2.1, 2 subjects in cohort-2.1, and 8 patients in cohort 5.1) was enrolled. 14 patients received talastatin in part 2 and part 5 dose studies (5 patients in part 2 cohort 2.1, 1 patient in part 2 cohort-2.1, and 8 patients in part 5 cohort 5.1). Both parts are administered stepwise. Specifically, for part 2 cohort 2.1, 1mg (the introduced dose) was administered to the patient on day 1 of cycle 1, then 20mg (the target dose) was administered on days 8 and 15 of cycle 1, after which 20mg of q3w was administered. For part 2 cohort-2.1, 1mg (the introduced dose) was administered to the patient on day 8 of cycle 1, followed by 20mg (the target dose) on day 15 of cycle 1, followed by 20mg of q3w. One subject in cohort-2.1 received chemotherapy and alemtuzumab on day 1 of cycle 1, but did not receive talatizumab treatment due to unrelated infection and ended the study. For part 5, 1mg (the introduced dose) was administered to the patient on day 1 of cycle 1, then 10mg (the target dose) was administered on days 8 and 15 of the cycle, after which 10mg q2w was administered. The alemtuzumab and chemotherapy are administered as described above.
For part 2, of the 5 subjects receiving talastatin in cohort 2.1, 5 subjects received imaging evaluation. 2 patients achieved confirmed Partial Remission (PR), 1 subject achieved unconfirmed PR, and 2 patients achieved Stable Disease (SD). One subject receiving talatizumab in cohort-2.1 reached unacknowledged PR. The total response of part 2 was 2/6 confirmed PR (33%), 2/6 unconfirmed PR (33%) and 2 cases SD (33%).
For part 5, of 8 subjects receiving talastatin, 5 subjects received an imaging evaluation. Of these 5 subjects, 3 reached SD (60%), and 2 had developed disease progression (40%). 3 subjects have not been subjected to imaging evaluation.
Since the high response rate to the initial standard treatment is known, the primary response assessment to the combination of talastatin with chemotherapy and PD-L1 inhibitor will be 6 months progression free and overall survival, and these measures are now evaluated as too early. Overall, to date, responses to the talastatin combination regimen are encouraging.
Among the 15 patients in the group, adverse events occurring in the treatment were observed in 14 (93.3%) patients. In 13 (86.7%) patients, a new occurrence of adverse events related to ≡2 treatment was observed. In cohort 2.1, part 2,2 subjects stopped treatment due to treatment-related adverse events. 1 subject experienced a grade 3 ICANS seizure event, and 1 subject experienced grade 4 thrombocytopenia associated with chemotherapy. Of the 15 subjects, 7 (46.7%) had developed cytokine release syndrome. 6 subjects experienced CRS grade 1, and 1 subject experienced CRS grade 2.
Table 11 below lists the sequences cited in the present application.
The present specification will be understood most thoroughly from the teachings of the references cited within the specification. The embodiments within the specification provide an illustration of embodiments of the invention and should not be construed as limiting the scope of the invention. The skilled person will readily recognize that many other embodiments are encompassed by the present invention. All publications, patents, and sequences cited in this disclosure are incorporated by reference in their entirety. In the event that a material incorporated by reference conflicts with or otherwise inconsistent with the present specification, the present specification will replace any such material. Citation of any reference herein is not an admission that such reference is prior art to the present invention.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following examples.

Claims (59)

1. A method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23, wherein the anti-DLL 3 agent is administered at a dose of from 10mg to 100mg twice every three weeks.
2. The method of claim 1, wherein the anti-DLL 3 agent is administered at a dose of 10mg, 30mg, or 100mg twice every three weeks.
3. The method of claim 1 or claim 2, wherein the anti-DLL 3 agent is administered on days 1 and 8 of a 21 day cycle.
4. A method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23, wherein the anti-DLL 3 agent is administered at a dose of from 20mg to 200mg once every three weeks.
5. The method of claim 4 wherein the anti-DLL 3 agent is administered at a dose of from 20mg to 100mg once every three weeks.
6. The method of claim 4 wherein the anti-DLL 3 agent is administered at a dose of from 100mg to 200mg once every three weeks.
7. The method of any one of claims 4-6 wherein the anti-DLL 3 agent is administered at a dose of 20mg, 60mg, 100mg, or 200 mg.
8. The method of any one of claims 4-7 wherein the anti-DLL 3 agent is administered on day 1 of a 21 day cycle.
9. A method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23, wherein the anti-DLL 3 agent is administered according to the following regimen:
a) Administering the anti-DLL 3 agent in a first period, wherein (i) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of from 1mg to 200mg over a period of 2 days to 7 days, and (ii) after the continuous intravenous infusion, the anti-DLL 3 agent is administered by bolus intravenous infusion on day 8, day 15, or both day 8 and day 15, and
B) Administering the anti-DLL 3 agent according to any one of the following i) to iii):
i) Starting on day 29 and once every two weeks thereafter, administering one or more subsequent doses of the anti-DLL 3 agent at a dose from 10mg to 100 mg;
ii) starting on day 22 and thereafter twice every three weeks, administering one or more subsequent doses of the anti-DLL 3 agent at a dose from 10mg to 100 mg; and
Iii) Starting on day 22 and once every three weeks thereafter, one or more subsequent doses of the anti-DLL 3 agent are administered at doses from 20mg to 200 mg.
10. The method of claim 9, wherein a) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of from 30mg to 100mg over a period of 2 days, 3 days, 5 days, or 7 days.
11. The method of claim 9 or 10, wherein a) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of 30mg, 50mg, or 100mg over a period of 2 days, 3 days, 5 days, or 7 days.
12. The method of any one of claims 9-11, wherein a) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of 30mg, 50mg, or 100mg over a period of 3 days, 5 days, or 7 days.
13. The method of claim 12, wherein a) the anti-DLL 3 agent is administered by continuous intravenous infusion at a dose of 30mg or 100mg over a period of 3 days.
14. A method of treating a DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23, an anti-PD-L1 antibody, and optionally one or more chemotherapeutic agents, wherein the anti-DLL 3 agent is administered according to any one of the following a) to c):
a) Administering the anti-DLL 3 agent at a dose of from 10mg to 100mg once every two weeks;
b) Administering the anti-DLL 3 agent twice every three weeks at a dose of from 10mg to 100 mg; and
C) The anti-DLL 3 agent is administered at a dose of from 20mg to 200mg once every three weeks.
15. The method of claim 14 wherein the anti-DLL 3 agent is administered once every two weeks at a dose of from 10mg to 100 mg.
16. The method of claim 15 wherein the anti-DLL 3 agent is administered at a dose of 10mg, 30mg, 50mg, or 100mg once every two weeks.
17. The method of claim 15 or 16 wherein the anti-DLL 3 agent is administered on days 1 and 15 of a 28 day cycle.
18. The method of claim 14 wherein the anti-DLL 3 agent is administered at a dose of from 10mg to 100mg twice every three weeks.
19. The method of claim 18, wherein the anti-DLL 3 agent is administered at a dose of 10mg, 30mg, or 100mg twice every three weeks.
20. The method of claim 18 or 19, wherein the anti-DLL 3 agent is administered on days 1 and 8 of a 21 day cycle.
21. The method of claim 14 wherein the anti-DLL 3 agent is administered once every three weeks at a dose of from 20mg to 100 mg.
22. The method of claim 14 wherein the anti-DLL 3 agent is administered once every three weeks at a dose of from 100mg to 200 mg.
23. The method of claim 21 or 22 wherein the anti-DLL 3 agent is administered at a dose of 20mg, 60mg, 100mg or 200mg once every three weeks.
24. The method of any one of claims 21-23 wherein the anti-DLL 3 agent is administered on day 1 of a 21 day cycle.
25. The method of any one of claims 14-24, wherein prior to any one of a) to c), the anti-DLL 3 agent is administered at a 21 day period according to the following regimen: a first dose of 0mg or 1mg on day 1, a second dose of from 1mg to 100mg on day 8, and a third dose of from 10mg to 200mg on day 15.
26. The method of claim 25, wherein the first dose is 1mg on day 1, the second dose is from 10mg to 100mg on day 8, and the third dose is from 10mg to 100mg on day 15.
27. The method of claim 25, wherein the first dose is 1mg on day 1, the second dose is from 10 to 100mg on day 8, and the third dose is from 20 to 200mg on day 15.
28. The method of any one of claims 14-24, wherein prior to any one of a) to c), the anti-DLL 3 agent is administered at a 21 day period according to the following regimen:
(i) A first dose of 1mg on day 1, a second dose of from 10mg to 100mg on day 8; or alternatively
(Ii) A first dose of 1mg on day 8 and a second dose of from 10mg to 100mg on day 15.
29. The method of claim 28, wherein the 21-day period is the first period of administration of the anti-DLL 3 to the subject, and wherein the second dose in (i) or (ii) is from 10mg to 50mg, preferably 10mg or 20mg.
30. The method of any one of claims 14-29, wherein the anti-PD-L1 antibody is a PD-L1 blocking antibody.
31. The method of claim 30, wherein the anti-PD-L1 antibody is alemtuzumab or devaluzumab.
32. The method of any one of claims 14-31, wherein the one or more chemotherapeutic agents comprise a platinum-based chemotherapeutic agent, etoposide, or both.
33. The method of claim 32, wherein the platinum-based chemotherapeutic agent is carboplatin or cisplatin.
34. The method of any one of claims 14-33, wherein the anti-DLL 3 agent is administered after administration of the anti-PD-L1 antibody and the one or more chemotherapeutic agents when administered on the same day.
35. A method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23 and an alkylating agent, wherein the anti-DLL 3 agent is administered to the subject at a dose of from 10mg to 200mg once every three weeks.
36. The method of claim 35 wherein the anti-DLL 3 agent is administered at a dose of from 10mg to 100mg once every three weeks.
37. The method of claim 36 wherein the anti-DLL 3 agent is administered at a dose of from 100mg to 200mg once every three weeks.
38. The method of any one of claims 35-37 wherein the anti-DLL 3 agent is administered at a dose of 10mg, 20mg, 60mg, 100mg, or 200 mg.
39. The method of any one of claims 35-38 wherein the anti-DLL 3 agent is administered on day 1 of a 21 day cycle.
40. A method of treating DLL3 positive cancer, the method comprising administering to a subject in need thereof an anti-DLL 3 agent comprising the amino acid sequences of SEQ ID NOs 13 and 23 and an alkylating agent, wherein the anti-DLL 3 agent is administered to the subject according to: a first dose of 0mg or 1mg on day 1, a second dose of from 10mg to 100mg on day 8, a third dose of from 10mg to 200mg on day 15, and one or more subsequent doses of from 10mg to 200mg starting on day 22 and once every three weeks thereafter.
41. The method of claim 40, wherein the first dose is 1mg, the second dose is from 10mg to 100mg, the third dose is from 10mg to 200mg, and the one or more subsequent doses are the same and the same as the third dose.
42. The method of claim 40, wherein the first dose is 1mg, the second dose is 20mg, 60mg, or 100mg, the third dose is 20mg, 60mg, or 200mg, and the one or more subsequent doses are the same and are each 20mg, 60mg, or 200mg.
43. The method of any one of claims 40-42, wherein the one or more subsequent doses are administered on day 1 of the 21-day cycle.
44. The method of any one of claims 35-43, wherein the alkylating agent is a platinum-based agent.
45. The method of claim 44, wherein the platinum-based agent is lubidine.
46. The method of claim 45, wherein the lubidine is administered once every three weeks at a dose of from 2.0mg/m 2 to 3.2mg/m 2.
47. The method of any one of claims 35-46 wherein the anti-DLL 3 agent is administered after administration of the alkylating agent when administered on the same day.
48. The method of any one of claims 1-47 wherein the anti-DLL 3 agent comprises the amino acid sequence of SEQ ID NO 14, 27 or 32.
49. The method of any one of claims 1-48, further comprising administering one or more additional therapeutic agents to the subject.
50. The method of claim 49, wherein the one or more additional therapeutic agents are corticosteroids, saline, or anti-IL 6 antibodies.
51. The method of claim 50, wherein the corticosteroid is dexamethasone.
52. The method of any one of claims 49-51 wherein the one or more additional therapeutic agents are administered in cycle 1 wherein the anti-DLL 3 agent is administered.
53. The method of any one of claims 1-52, wherein the cancer is Small Cell Lung Cancer (SCLC).
54. The method of any one of claims 1-53, wherein the cancer is recurrent/refractory SCLC (RR SCLC) or extensive disease SCLC (ED SCLC).
55. The method of any one of claims 1-54, wherein the subject is a human.
56. The method of claim 55, wherein the subject has received at least one prior treatment for the cancer and has relapsed.
57. The method of claim 56, wherein the at least one prior treatment for the cancer is platinum chemotherapy, etoposide, and optionally an anti-PD-L1 antibody.
58. The method of claim 55, wherein the subject has not received prior systemic treatment for the cancer.
59. The method of any one of claims 1-58 wherein the anti-DLL 3 agent is talatizumab.
CN202380022555.3A 2022-02-23 2023-02-22 DLL3 targeted cancer treatment Pending CN118785922A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/313,119 2022-02-23
US202263429311P 2022-12-01 2022-12-01
US63/429,311 2022-12-01
PCT/US2023/063010 WO2023164474A1 (en) 2022-02-23 2023-02-22 Cancer treatment targeting dll3

Publications (1)

Publication Number Publication Date
CN118785922A true CN118785922A (en) 2024-10-15

Family

ID=92997046

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202380022555.3A Pending CN118785922A (en) 2022-02-23 2023-02-22 DLL3 targeted cancer treatment

Country Status (1)

Country Link
CN (1) CN118785922A (en)

Similar Documents

Publication Publication Date Title
US11866509B2 (en) Humanized antibodies against CEACAM1
KR102702287B1 (en) Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
JP2022068352A (en) Treatment method of lung cancer using combination of anti-pd-1 antibody and anti-ctla-4 antibody
KR20190015407A (en) Anti-PD-1 antibody for use in the treatment of recurrent small cell lung cancer
JP2024038034A (en) Anti-pd-l1 antibody treatment of bladder cancer
WO2020211804A1 (en) Use of anti-pd-1 antibody in preparation of medicament for treating solid tumors
US20240317856A1 (en) Dosing regimen for combination therapy targeting dll3 and pd-1
AU2019240200B2 (en) Pharmaceutical combinations
CN118785922A (en) DLL3 targeted cancer treatment
US11427647B2 (en) Polynucleotides encoding humanized antibodies against CEACAM1
TW202309078A (en) Methods and compositions for treating cancer
JP2024511977A (en) Cancer treatment method using anti-ILT3 antibody
KR20240153575A (en) Cancer treatments targeting DLL3
US20240092934A1 (en) Assessment of ceacam1 expression on tumor infiltrating lymphocytes
US20240376206A1 (en) Compositions and methods for treating late stage lung cancer
TW202436335A (en) Methods for treating dll3-expressing cancer
KR20240038204A (en) Composition for preventing or treating neuroendocrine neoplasm comprising an antibody that specifically binds to pd-1 as an active ingredient
CN117940452A (en) Methods and compositions for treating cancer
CN118251234A (en) Anti-galectin-9 antibodies and therapeutic uses thereof
CN113164599A (en) Use of anti-PD-L1 monoclonal antibody for treating cancer
EA045953B1 (en) TREATING BLADDER CANCER WITH PD-L1 ANTIBODY

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication