WO2019178438A1 - Abbv-621 in combination with anti-cancer agents for the treatment of cancer - Google Patents

Abbv-621 in combination with anti-cancer agents for the treatment of cancer Download PDF

Info

Publication number
WO2019178438A1
WO2019178438A1 PCT/US2019/022398 US2019022398W WO2019178438A1 WO 2019178438 A1 WO2019178438 A1 WO 2019178438A1 US 2019022398 W US2019022398 W US 2019022398W WO 2019178438 A1 WO2019178438 A1 WO 2019178438A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
abbv
subject
irinotecan
combination
Prior art date
Application number
PCT/US2019/022398
Other languages
French (fr)
Inventor
Susan E. Morgan-Lappe
Darren C. Phillips
Fritz G. Buchanan
Xin Lu
Monica Motwani
Original Assignee
Abbvie 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 Abbvie Inc. filed Critical Abbvie Inc.
Publication of WO2019178438A1 publication Critical patent/WO2019178438A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Abstract

This invention pertains to a method for the treatment of cancer in a subject comprising administering to the subject an effective amount of ABBV-621 in combination with anti-cancer agents.

Description

ABBV-621 IN COMBINATION WITH ANTI-CANCER AGENTS FOR THE
TREATMENT OF CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Serial No.
62/643,242, filed March 15, 2018; and claims the benefit of U.S. Provisional Application Serial No. 62/815,788, filed March 8, 2019; the disclosures of which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
[0001] The present invention pertains to a method for the treatment of cancer in a subject comprising administering to the subject an effective amount of ABBV-621 in combination with anti-cancer agents.
BACKGROUND OF THE INVENTION
[0002] Cell death can be initiated through activation of the extrinsic and intrinsic apoptotic signalling pathways. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily of cytokines, preferentially triggers the extrinsic apoptotic pathway by binding as a trimer to two closely related cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5). When these receptors undergo trimerization, it leads to the formation of the death-inducing signalling complex (DISC) to recruit and activate downstream caspases that ultimately leads to apoptotic cell death. Since TRAIL was found to induce apoptosis, several TRAIL receptor agonists have been developed for the treatment of cancer.
[0003] ABBV-621 is a novel, second generation TRAIL receptor agonist that is currently being tested in Phase 1 clinical trials. It is an engineered fusion protein consisting of an IgGl-Fc linked to a single chain trimer of TRAIL subunits resulting in a total of six death receptor binding sites per molecule in order to maximize receptor clustering. ABBV-621 exhibited potent single agent activity in acute myeloid lymphoma (AML) and diffuse large B-cell lymphoma (DLBCL) cell lines following 24hrs treatment. This activity was rapid and mechanism-based since activation of downstream apoptotic signalling events (caspase activation, mitochondrial depolarization, phosphatidylserine exposure) were observed as early as 1 hour following the addition of ABBV- 621, and cell death could be completely blocked with the pan caspase inhibitor z-VAD-FMK.
[0004] The activity of ABBV-621 has been established in numerous cancer types both in vitro and in vivo and is potent as a single agent. Specifically, ABBV-621 has demonstrated single agent anti-tumor activity in AML and DLBCL in in vivo models. However, certain cell types are resistant to ABBV-621 as a single agent. The current disclosure demonstrates for the first time the ability of ABBV-621 to synergize with several standard of care anti-cancer agents (paclitaxel, irinotecan, FOLFIRI, bortezomib, and docetaxel).
BRIEF SUMMARY OF THE INVENTION
[0005] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents.
[0006] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, lung cancer, non-small cell lung cancer, colorectal cancer, and gastric cancer.
[0007] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and further wherein the anti-cancer agent is selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, the combination of FOLFIRI and bevacizumab, bortezomib, and docetaxel.
[0008] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, the combination of FOLFIRI and bevacizumab, bortezomib, and docetaxel, and further wherein the anti-cancer agent is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0009] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is bortezomib which is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0010] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is paclitaxel which is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0011] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is irinotecan which is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0012] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is FOLFIRI which is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0013] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent comprises the combination of FOLFIRI and bevacizumab, which anti-cancer agent is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0014] The present invention pertains to a method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, wherein the anti-cancer agent is docetaxel which is administered either prior to, subsequent to, or concurrently with ABBV-621.
[0015] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, the combination of FOLFIRI and bevacizumab, bortezomib, and docetaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone.
[0016] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, combination of FOLFIRI and bevacizumab, bortezomib, and docetaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, and wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer.
[0017] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, combination of FOLFIRI and bevacizumab, bortezomib, and docetaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the anti-cancer agent is administered prior to, subsequent to, or concurrently with ABBV-621.
[0018] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with paclitaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the paclitaxel is administered prior to, subsequent to, or concurrently with ABBV-621.
[0019] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with bortezomib, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the bortezomib is administered prior to, subsequent to, or concurrently with ABBV-621.
[0020] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with irinotecan, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the irinotecan is administered prior to, subsequent to, or concurrently with ABBV-621.
[0021] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with FOLFIRI, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the FOLFIRI is administered prior to, subsequent to, or concurrently with ABBV-621.
[0022] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with FOLFIRI and/or bevacizumab, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the FOLFIRI and/or bevacizumab is administered prior to, subsequent to, or concurrently with ABBV-621.
[0023] The present invention pertains to method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with docetaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer, and wherein the docetaxel is administered prior to, subsequent to, or concurrently with ABBV-621.
BRIEF DESCRIPTION OF THE DRAWINGS [0024] Figure 1 shows the growth inhibition curves of xenografted human colorectal adenocarcinoma tumors (DLD-l) by ABBV-621 in combination with Irinotecan as described in Example 1. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0025] Figure 2 shows the growth inhibition curves growth inhibition of xenografted human multiple myeloma tumors (OPM-2) by ABBV-621 in combination with Bortezomib as described in Example 2. As shown, the combination of ABBV-621 with Bortezomib resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0026] Figure 3 shows the growth inhibition curves growth inhibition of xenografted human non-small cell lung carcinoma tumors (H460-LM) by ABBV-621 in combination with Paclitaxel as described in Example 3. As shown, the combination of ABBV-621 with Paclitaxel resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0027] Figure 4 shows the growth inhibition curves growth inhibition of xenografted human colorectal carcinoma tumors (SW-48) by ABBV-621 in combination with Irinotecan as described in Example 4. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0028] Figure 5 shows the growth inhibition curves of xenografted human gastric carcinoma tumors (Hs746t) by ABBV-621 in combination with Irinotecan as described in Example 5. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0029] Figure 6 shows the growth inhibition curves of xenografted human gastric carcinoma tumors (Hs746t) by ABBV-621 in combination with Docetaxel as described in Example 6. As shown, the combination of ABBV-621 with Docetaxel resulted in enhanced tumor growth inhibition relative to either treatment alone. [0030] Figure 7 shows the growth inhibition curves of xenografted human gastric carcinoma tumors (NCI-N87) by ABBV-621 in combination with Irinotecan as described in Example 7. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0031] Figure 8 shows the growth inhibition curves of xenografted human gastric carcinoma tumors (NCI-N87) by ABBV-621 in combination with Docetaxel as described in Example 8. As shown, the combination of ABBV-621 with Docetaxel resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0032] Figure 9 shows higher expression levels for DR4 and DR5 RNA expression in tumors with a KRAS mutation compared to WT (colorectal cancer tumors).
[0033] Figure 10 shows higher expression levels for DR4 and DR5 RNA expression in tumors with a KRAS mutation compared to WT (various cancer tumors).
[0034] Figure 11 shows the growth inhibition curves of xenografted human Colorectal carcinoma tumors (Colo-205) by ABBV-621 in combination with Irinotecan as described in Example 1. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0035] Figure 12 shows the growth inhibition curves of xenografted human colorectal carcinoma tumors (SW-48) by ABBV-621 in combination with Irinotecan as described in Example 11. As shown, the combination of ABBV-621 with irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
[0036] Figure 13 shows the growth inhibition curves of patient derived xenografted human colorectal carcinoma tumors (CTG-0064) by ABBV-621 in combination with Irinotecan as described in Example 11. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone. [0037] Figure 14 shows the growth inhibition curves of patient derived xenografted human colorectal carcinoma tumors (CTG-0069) by ABBV-621 in combination with Irinotecan as described in Example 11. As shown, the combination of ABBV-621 with Irinotecan resulted in enhanced tumor growth inhibition relative to either treatment alone.
DETAILED DESCRIPTION OF THE INVENTION
[0038] The activity ABBV-621 has been established in numerous cancer types both in vitro and in vivo and is potent as a single agent. However, certain cell types are resistant to ABBV-621 as a single agent. The present inventors demonstrate, for the first time, that ABBV-621 synergizes, in vivo, with a variety of standard of care cancer agents (irinotecan, bortezomib, paclitaxel, and docetaxel) in xenograft cancer models (multiple myeloma, non-small cell lung carcinoma, colorectal carcinoma, and gastric cancer) which support the use of ABBV-621 for treating these cancers when used in combination with one or more of these agents.
[0039] In addition, the present inventors also demonstrate for the first time that patient derived xenograft models containing KRAS mutations had higher anti-tumor responses to single agent ABBV-621 compared with patient derived xenograft models containing wild type KRAS. Importantly, the inventors also discovered that DR4 and DR5 mRNA levels were higher in KRAS mutant (gain of function) tumors compared to KRAS wild-type tumors. Accordingly, KRAS mutations either alone or in conjunction with higher levels of death receptors may serve as a biomarker for predicting patients that have a higher probability of achieving increased sensitivity and/or response to ABBV-621.
[0040] In one embodiment, the present invention relates to ABBV-621, a TNF-related apoptosis inducing ligand (TRAIL) receptor agonist protein. It is disclosed as a TRAIL receptor agonist protein comprising a dimer of two polypeptides having the amino acid sequence set forth in SEQ ID NO: 1, wherein the two polypeptides are covalently linked through three interchain disulphide bonds formed between cysteine residues 513, 519 and 522 of each polypeptide, and wherein one or more of the asparagine residues at postions 168 and 337 of the polypeptide(s) are N- glycosylated, and further post-translationally modifed by changing the A-termian glutamine to pyroglutamate, as disclosed in US Patent Publication No. 2015/0337027, incorporated herein by reference in its entirety and for all purposes.
[0041] As is known in the art, the therapeutic form of irinotecan is irinotecan hydrochloride (referred to herein as Compound (I)) for injection and has a CAS Registry Number of 100286-90- 6; has an empirical formula of C33H38N406*HCl»3H20; a gram molecular weight is 677.19; and is described in U.S. Patent No. 4,604,463, published August 5, 1986, incorporated herein by reference in its entirety and for all purposes. Use of the term "(S)-4, l l-diethyl-3,4,l2,l4- tetrahydro-4-hydroxy-3, l4-dioxolHpyrano[3’,4’:6,7]-indolizino[l,2-b]quinolin-9-yl- [l,4’bipiperidine]-l’-carboxylate, monohydrochloride, trihydrate" encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of irinotecan or its salts. Pharmaceutical compositions of (S)-4,l l-diethyl-3,4,l2, l4-tetrahydro-4-hydroxy-3, l4- dioxolHpyrano[3,,4, :6,7]-indolizino[l,2-b]quinolin-9-yl-[l,4,bipiperidine]-l’-carboxylate, monohydrochloride, trihydrate include all pharmaceutically acceptable compositions comprising (S)-4,l l-diethyl-3,4, l2,l4-tetrahydro-4-hydroxy-3,l4-dioxolHpyrano[3’,4’:6,7]-indolizino[l,2- b]quinolin-9-yl-[l,4’bipiperidine]-l’-carboxylate, monohydrochloride, trihydrate and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising (S)-4,l l-diethyl-3,4, l2,l4-tetrahydro-4-hydroxy-3,l4-dioxolHpyrano[3’,4’:6,7]-indolizino[l,2- b]quinolin-9-yl-[l,4,bipiperidine]-l’-carboxylate, monohydrochloride, trihydrate is CAMPTOSAR® (Upjohn). CAMPTOSAR® comprises (S)-4, l l-diethyl-3,4,l2, l4-tetrahydro-4- hydroxy-Srid-dioxolHpyranofS’^kb j-indolizinof l^-bjquinolin^-yl-f kd’bipiperidine]-!’- carboxylate, monohydrochloride, trihydrate as the active ingredient, also referred to as irinotecan hydrochloride for injection. CAMPTOSAR® is supplied as a sterile, pale yellow, clear, aqueous solution. Each milliliter of solution contains 20 mg of irinotecan hydrochloride (on the basis of the trihydrate salt), 45 mg of sorbitol, NF, and 0.9 mg of lactic acid, USP. The pH of the solution has been adjusted to 3.5 (range, 3.0 to 3.8) with sodium hydroxide or hydrochloric acid. CAMPTOSAR® is intended for dilution with 5% Dextrose Injection, USP (D5W), or 0.9% Sodium Chloride Injection, USP, prior to intravenous infusion. The preferred diluent is 5% Dextrose Injection, USP. Another example of a pharmaceutical composition comprising“(45)-4, l l-diethyl- 4-hydroxy-3, l4-dioxo-3,4, l2,l4-tetrahydro-liT-pyrano[3',4':6,7]indolizino[l,2-Z>]quinolin-9-yl [l,4'-bipiperidine]-T-carboxylate is ONIVYDE™ (Merrimack Pharmaceuticals, Inc ). ONIVYDE is a sterile, white to slightly yellow opaque isotonic liposomal dispersion. Each 10 mL single-dose vial contains 43 mg irinotecan free base at a concentration of 4.3 mg/mL. The liposome is a unilamellar lipid bilayer vesicle, approximately 110 nm in diameter, which encapsulates an aqueous space containing irinotecan in a gelated or precipitated state as the sucrose octasulfate salt. The vesicle is composed of l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) 6.81 mg/mL, cholesterol 2.22 mg/mL, and methoxy -terminated polyethylene glycol (MW 2000)- distearoylphosphatidyl ethanolamine (MPEG-2000-DSPE) 0.12 mg/mL. Each mL also contains 2- [4-(2 -hydroxy ethyl) piperazin-l-yl]ethanesulfonic acid (HEPES) as a buffer 4.05 mg/mL and sodium chloride as an isotonicity reagent 8.42 mg/mL.
Figure imgf000013_0001
Compound (I)
[0042] As is known in the art, the therapeutic form of bortezomib is bortezomib monomeric boronic acid for injection (referred to herein as Compound (II)) and has a CAS Registry Number of 179324-69-7; has an empirical formula of C19H25BN4O4; a gram molecular weight is 384.24; and is described in U.S. Patent No. US 5,780,454, published July 14, 1998, incorporated herein by reference in its entirety and for all purposes. Use of the term "[(lR)-3-methyl-l-[[(2S)-l-oxo-3- phenyl-2-[(pyrazinylcarbonyl)-amino]propyl]amino]butyl]-boronic acid" encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of bortezomib or its salts. Pharmaceutical compositions of [(lR)-3-methyl-l-[[(2S)-l-oxo-3-phenyl-2- [(pyrazinylcarbonyl)-amino]propyl]amino]butyl]-boronic acid include all pharmaceutically acceptable compositions comprising [(lR)-3-methyl-l-[[(2S)-l-oxo-3-phenyl-2- [(pyrazinylcarbonyl)-amino]propyl]amino]butyl]-boronic acid and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising [(lR)-3-methyl-l- [[(2S)-l-oxo-3-phenyl-2-[(pyrazinylcarbonyl)-amino]propyl]amino]butyl]-boronic acid is VELCADE® (Millenium). VELCADE® comprises [(lR)-3-methyl-l-[[(2S)-l-oxo-3-phenyl-2- [(pyrazinylcarbonyl)-amino]propyl]amino]butyl]-boronic acid as the active ingredient, also referred to as bortezomib monomeric boronic acid for injection. VELCADE® is supplied as a sterile, pale yellow, clear, aqueous solution. Each vial contains 3.5 mg of bortezomib as a sterile lyophilized powder, and 35 mg mannitol, USP, as an inactive ingredient. VELCADE® is a mannitol boronic ester which, in reconstituted form, consists of the mannitol ester in equilibrium with its hydrolysis product, the monomeric boronic acid. The drug substance exists in its cyclic anhydride form as a trimeric boroxine.
Figure imgf000014_0001
Compound (II)
[0043] As is known in the art, the therapeutic form of paclitaxel is paclitaxel for injection (referred to herein as Compound (III)) and has a CAS Registry Number of 33069-62-4; has an empirical formula of C47H51NO14; a gram molecular weight is 853.9; and is described in M. C. Wani et al., J. Am. Chem. Soc. 93, 2325 (1971), published in May 1971, incorporated herein by reference in its entirety and for all purposes. Use of the term "5b, 20-Epoxy- 1 ,2a,4,7b, 10b, 13a- hexahydroxytax-l l-en-9-one 4,10-diacetate 2-benzoate l3-ester with (2R,3S)-N-benzoyl-3- phenylisoserine" encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of paclitaxel or its salts. Pharmaceutical compositions of 5b,20-Epoxy- l,2a,4,7b,10b,13a-hexahydroxytax-l l-en-9-one 4,10-diacetate 2-benzoate l3-ester with (2R,3S)- N-benzoyl-3-phenylisoserine include all pharmaceutically acceptable compositions comprising 5b,20-Epoxy-l,2a,4,7b,10b,13a-hexahydroxytax-l l-en-9-one 4,10-diacetate 2-benzoate l3-ester with (2R,3S)-N-benzoyl-3-phenylisoserine and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising 5b,20-Epoxy-l,2a,4,7b,10b,13a- hexahydroxytax-l l-en-9-one 4,10-diacetate 2-benzoate l3-ester with (2R,3S)-N-benzoyl-3- phenylisoserine is TAXOL® (Bristol-Myers Squibb). TAXOL® comprises 5b,20-Erocg- 1 ,2a,4,7b, 10b, 13a-hexahydroxytax- 1 1 -en-9-one 4,10-diacetate 2-benzoate l3-ester with (2R,3S)- N-benzoyl-3-phenylisoserine as the active ingredient, also referred to as paclitaxel for injection. TAXOL® is supplied as a clear, colorless to slightly yellow viscous, nonaqueous solution, intended for dilution with a suitable parenteral fluid prior to intravenous infusion. Each vial contains sterile nonpyrogenic solution contains 6 mg paclitaxel, 527 mg of purified Cremophor® EL* (polyoxy ethylated castor oil) and 49.7% (v/v) dehydrated alcohol, LISP. Paclitaxel is also available as an alternative formulation, ABRAXANE® for Injectable Suspension (Celgene Corporation) (paclitaxel protein-bound particles for injectable suspension, nab-paclitaxel) (albumin-bound) is an albumin-bound form of paclitaxel with a mean particle size of approximately 130 nanometers. Paclitaxel exists in the particles in a non-crystalline, amorphous state. ABRAXANE® is supplied as a white to yellow, sterile, lyophilized powder for reconstitution with 20 mL of 0.9% Sodium Chloride Injection, LISP prior to intravenous infusion. Each single-use vial contains 100 mg of paclitaxel (bound to human albumin) and approximately 900 mg of human albumin (containing sodium caprylate and sodium acetyltryptophanate). Each milliliter (mL) of reconstituted suspension contains 5 mg paclitaxel. ABRAXANE® is free of solvents.
Figure imgf000015_0001
Compound (III) [0044] As is known in the art, the therapeutic form of docetaxel is docetaxel inj ection concentrate (referred to herein as Compound (IV)) and has a CAS Registry Number of 114977-28-5; has an empirical formula of C43H53NO14· 3H20; a gram molecular weight is 861.9; and is described in U.S. Patent No. US 4,814,470, published March 21, 1989, incorporated herein by reference in its entirety and for all purposes. Use of the term (2R,3S)-N-carboxy-3-phenylisoserine, N-tert-butyl ester, l3-ester with 5b-20-erocn- 1 ,2a,4,7b, 10b, 13a-hexahydroxytax- 1 1 -en-9-one 4-acetate 2- benzoate, trihydrate" encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of docetaxel or its salts. Pharmaceutical compositions of (2R,3S)-N-carboxy- 3-phenylisoserine, N-tert-butyl ester, l3-ester with 5b-20-epoxy-l,2a,4,7b,10b,13a- hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate include all pharmaceutically acceptable compositions comprising (2R,3S)-N-carboxy-3-phenylisoserine, N-tert-butyl ester, 13- ester with 5b-20-epoxy-l,2a,4,7b,10b, 13a-hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising (2R,3S)-N-carboxy-3-phenylisoserine, N-tert-butyl ester, l3-ester with 5b-20-epoxy-l,2a,4,7b,10b,13a-hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate is TAXOTERE® (RPR) Injection Concentrate. TAXOTERE ® comprises (2R,3S)-N-carboxy-3- phenylisoserine, N-tert-butyl ester, l3-ester with 5b-20-epoxy-l,2a,4,7b,10b,13a- hexahydroxytax-l l-en-9-one 4-acetate 2-benzoate, trihydrate as the active ingredient, also referred to as docetaxel for injection. TAXOTERE ® is supplied as a clear yellow to brownish- yellow viscous, non-progenic, sterile solution. Each vial contains 20 mg (0.5 mL) or 80 mg (2 mL) docetaxel (anhydrous). Each mL contains 40 mg docetaxel (anhydrous) and 1040 mg polysorbate 80. TAXOTERE Injection Concentrate requires dilution with Diluent prior to addition to the infusion bag. The diluent for TAXOTERE contains 13% ethanol in water for injection and is supplied in vials.
Figure imgf000017_0001
Compound (IV)
[0045] As is known in the art, FOLFIRI is a chemotherapeutic regimen comprising folinic acid (calcium folinate), 5-fluorouracil, and irinotecan.
[0046] As is known in the art, the therapeutic form of bevacizumab is bevacizumab injection and has a CAS Registry Number of 216974-75-3; has an empirical formula of C6538H10034N1716O2033S44; and has an approximate molecular weight of 149 kDa. Bevacizumab is comprised of the heavy chain protein binding sequence of SEQ ID NO: 3 and the light chain protein binding sequence of SEQ ID NO: 2. Bevacizumab is vascular endothelial growth factor directed antibody. Bevacizumab is a recombinant humanized monoclonal IgGl antibody that contains human framework regions and murine complementarity-determining regions. Bevacizumab is indicated for the treatment of patients with metastatic colorectal cancer, non-squamous non-small cell lung cancer, glioblastoma, cervical cancer, epithelial ovarian, fallopian tube or primary peritoneal cancer. One example of a pharmaceutical composition comprising bevacizumab is AVASTIN® Injection. In one embodiment, the present invention relates to a method for the treatment of adenocarcinoma cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with irinotecan.
[0047] In one embodiment, the present invention relates to a method for the treatment of adenocarcinoma cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with FOLFIRI. [0048] In one embodiment, the present invention relates to a method for the treatment of multiple myeloma cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with bortezomib.
[0049] In one embodiment, the present invention relates to a method for the treatment of non small cell lung cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with paclitaxel.
[0050] In one embodiment, the present invention relates to a method for the treatment of gastric cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with irinotecan.
[0051] In one embodiment, the present invention relates to a method for the treatment of gastric cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with docetaxel.
[0052] In one embodiment, the present invention relates to a method for the treatment of multiple myeloma, non-small cell lung carcinoma, colorectal carcinoma, and gastric cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent.
[0053] In one embodiment, the present invention relates to a method for the treatment of adenocarcinoma cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, Compound (I).
[0054] In one embodiment, the present invention relates to a method for the treatment of colorectal cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, Compound (I).
[0055] In one embodiment, the present invention relates to a method for the treatment of gastric cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, Compound (I). [0056] In one embodiment, the present invention relates to a method for the treatment of colorectal cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, FOLFIRI.
[0057] In one embodiment, the present invention relates to a method for the treatment of KRAS mutant positive colorectal cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI.
[0058] In one embodiment, the present invention relates to a method for the treatment of colorectal cancer in a subject that has elevated DR4 and/or DR5 expression levels, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI.
[0059] In one embodiment, the present invention relates to a method for the treatment of KRAS mutant positive colorectal cancer in a subject that also has elevated DR4 and/or DR5 expression levels, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI.
[0060] In one embodiment, the present invention relates to a method for the treatment of colorectal cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, FOLFIRI and/or bevacizumab.
[0061] In one embodiment, the present invention relates to a method for the treatment of KRAS mutant positive colorectal cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI and/or bevacizumab.
[0062] In one embodiment, the present invention relates to a method for the treatment of colorectal cancer in a subject that has elevated DR4 and/or DR5 expression levels, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI and/or bevacizumab. [0063] In one embodiment, the present invention relates to a method for the treatment of KRAS mutant positive colorectal cancer in a subject that also has elevated DR4 and/or DR5 expression levels, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, FOLFIRI and/or bevacizumab.
[0064] In one embodiment, the present invention relates to a method for the treatment of multiple myeloma cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, Compound (II).
[0065] In one embodiment, the present invention relates to a method for the treatment of non small cell lung cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti cancer agent, Compound (III).
[0066] In one embodiment, the present invention relates to a method for the treatment of gastric cancer in a subject who is in need thereof, comprising administering to the subject an effective amount of the TRAIL receptor agonist, ABBV-621, with an apoptosis inducing anti-cancer agent, Compound (IV).
[0067] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the TRAIL receptor agonist ABBV-621 or a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of:
irinotecan, FOLFIRI, or FOLFIRI and bevacizumab, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone.
[0068] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the TRAIL receptor agonist ABBV-621. [0069] In one embodiment, the present invention relates to a method for the treatment of KRAS mutant positive cancer in a subject comprising the administration of a synergistic, therapeutically effective amount of the TRAIL receptor agonist
[0070] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with an anti-cancer agent, irinotecan.
[0071] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with an anti-cancer agent, FOLFIRI.
[0072] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with an anti-cancer agent, FOLFIRI, and bevacizumab.
[0073] In one embodiment, the present invention relates to a method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with an anti-cancer agent, docetaxel. In one embodiment, the present invention relates to a synergistic method for the treatment of cancer in a subject who is in need thereof, comprising administering to the subject one or more anti-cancer agents selected from Compound (I), Compound (II), Compound (III), Compound (IV), FOLFIRI, and FOLFIRI in combination with bevacizumab prior to administering an effective amount of the TRAIL receptor agonist, ABBV-621, such that said anti-cancer agent is administered for a sufficient period of time before ABBV-621 to ensure the subject is able to achieve either synergistic or optimal synergistic efficacy, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer. In this context,“sufficient time” may be construed to mean about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours ± about 15 to about 45 minutes. [0074] Compound (I), Compound (II), Compound (III), Compound (IV) FOLFIRI, and FOLFIRI in combination with bevacizumab includes all pharmaceutically acceptable compositions and one or more diluents, vehicles and/or excipients.
[0075] KRAS mutation status is most commonly associated with predicting patient response to EGFR inhibitors. For example, KRAS mutation status can predict the response to the anti-EGFR monoclonal antibodies cetuximab and panitumumab (Allegra, 2009). Tumors harboring activating mutations of KRAS, a signaling molecule downstream of EGFR, do no benefit from anti-EGFR therapy (Linardou, 2008). In particular, the most common activating KRAS mutations reside in codons 12, 13, and 61 of the KRAS gene are confer constitutive growth signaling and hence resistance to compounds targeting the EGFR signaling pathway (Andre, T., et al., Annals of Oncology, 2012, 00: 1-8; DeRook, W., et al., Lancit Oncology, 2010, 11 : 6753- 6762). In KRAS wild type (WT) patients, on the other hand, the addition of cetuximab to cytotoxic treatment improves the response rates with 16 to 24% compared to cytotoxic therapy alone. About 40% of the KRAS WT patients are non-responders to combination therapy
(Bokemeyer, 2009; Van Cutsem, 2009) and a significantly larger fraction of patients are non responders to EGFR antibody monotherapy (Amado, 2008).
[0076] Surprisingly, the present inventors have discovered for the first time that patient derived xenograft animal models that are KRAS mutation positive had an increased therapeutic response to therapeutic treatment with ABBV-621. By implication, colorectal cancer patients that are deemed to be KRAS mutant positive may have an increased likelihood of achieving a favorable therapeutic response to the administration of ABBV-621, and/or the combinations outlined herein.
[0077] The present invention contemplates a number of methods for detecting the presence of a KRAS mutation in a patient’s tumor tissue, including allele-specific PCR, real-time PCR methods with melt-curve analysis, high-resolution melting tests (see ET.S. Patent No. 8,980,556); and nucleic acid sequencing techniques. In one embodiment, the KRAS detection method is the THERASCREEN® KRAS RGQ PCR assay (Qiagen) or the cobas® KRAS Mutation Test (Roche). [0078] As used herein, the terms "KRAS" and“k-ras” are used interchangeably and refer to the human cellular homolog of a transforming gene isolated from the Kirsten rat sarcoma virus, as defined by NCBI's OMIM database entry 190070, and/or its expression products.
[0079] The phrases“KRAS mutant positive patient” and“KRAS mutant positive colorectal cancer patient” refers to a patient that has been confirmed to harbor one or more activating KRAS mutations known in the art, and is used herein to serve as a genetic signature or patient profile that can be used as an objective means of classifying patients whom may have a higher likelihood of achieving a favorable therapeutic response to the administration of ABBV-621, the combination of ABBV-621 and irinotecan, the combination of ABBV-621 and FOLFIRI, the combination of ABBV-621, FOLFIRI, and bevacizumab, and/or other combinations outlined herein. The most common activating KRAS mutations reside in codons 12, 13, 61, and 146 of the KRAS gene which encode one or more of the following, non-limiting mutations in the encoded KRAS protein: G12D; G12V; G12C; G12A; G12S; G12R; G12F; G13V; G13D; G13C; G13S; G13R; G13A; G13D; Q61H; Q61L; Q61H; Q61R; Q61K; and A146T.
[0080] As described herein, KRAS mutation positive status may be used as part of a therapeutic treatment regimen as a means of identifying those patients that have the highest likelihood of achieving a favorable therapeutic response to ABBV-621 and combinations described herein. In some embodiments, the method of treatment may involve assessing whether a colorectal cancer patient is KRAS mutant positive by detecting the presence of one or more KRAS mutations in a biological sample from said patient followed by the administration of ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab, etc.) if said patient is deemed to be KRAS mutant positive.
[0081] In one aspect, the present invention provides a method of treating a colorectal patient comprising the step of determining whether said patient is KRAS mutant positive by testing a biological sample from said patient using a PCR detection method, and if said patient is determined to be KRAS mutant positive, administering ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab, etc.), wherein said detection method is the THERASCREEN® KRAS RGQ PCR assay (Qiagen). [0082] In one aspect, the present invention provides a method of treating a colorectal patient comprising the step of determining whether said patient is KRAS mutant positive by testing a biological sample from said patient using a PCR detection method, and if said patient is determined to be KRAS mutant positive, administering ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab, etc.), wherein said detection method is the cobas® KRAS Mutation Test (Roche).
[0083] In one aspect, the present invention provides a method of treating a colorectal patient comprising the step of determining whether said patient is KRAS mutant positive by testing a biological sample from said patient using a KRAS mutant detection method, and administering ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab, etc.), wherein said KRAS mutant detection method is selected from the group consisting of: allele-specific PCR, real-time PCR methods with melt-curve analysis, high- resolution melting tests; and nucleic acid sequencing techniques.
[0084] In another aspect, elevated expression of either DR4 and/or DR5 in a patient tissue sample, as determined by comparing the level of DR4 and/or DR5 expression to a reference standard, may be used either alone or in conjunction with KRAS mutant positive status as an additional means of identifying patients that have a higher likelihood of achieving a favorable therapeutic response to therapies comprising ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab;
FOLFIRI and bevacizumab; and irinotecan and bevacizumab , etc.). The reference standard may be determined by comparing DR4 and/or DR5 to standard housekeeping gene expression to obtain a relative expression level and comparing that relative expression level to a standard level of expression for DR4 and/or DR5 and assessing whether or not DR4 and/or DR5 are elevated.
[0085] In one aspect, the present invention provides a method of treating a colorectal patient comprising the step of determining whether said patient has elevated expression of either DR4, DR5, and/or both DR4 and DR5, by measuring the expression level of said one or more receptor from a biological sample from said patient, and if the patient is deemed to have elevated expression in one or more of these receptors, administering ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab , etc.).
[0086] In one aspect, the present invention provides a method of treating a colorectal patient comprising the step of determining whether said patient has elevated expression of either DR4, DR5, and/or both DR4 and DR5, in addition to determining whether said patient is KRAS mutant positive, by measuring the expression level of said one or more receptor from a biological sample from said patient, as well as detecting whether said patient is KRAS mutant positive, and if the patient is deemed to have elevated expression in one or more of these receptors in addition to being KRAS mutant positive, administering ABBV-621 and/or ABBV-621 in combination with one or more therapeutic agents described herein (e.g., irinotecan; FOLFIRI; bevacizumab; FOLFIRI and bevacizumab; and irinotecan and bevacizumab , etc.).
[0087] Unless otherwise defined herein, scientific and technical terms used herein have the meanings that are commonly understood by those of ordinary skill in the art. In the event of any latent ambiguity, definitions provided herein take precedent over any dictionary or extrinsic definition. Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. The use of“or” means“and/or” unless stated otherwise. The use of the term“including”, as well as other forms, such as“includes” and“included”, is not limiting. Any range described here will be understood to include the endpoints and all values between the endpoints.
[0088] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference in their entirety for any purpose. To the extent documents incorporated by reference contradict the disclosure contained in the specification; the specification will supersede any contradictory material.
[0089] Generally, nomenclatures used in connection with cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are those well-known and commonly used in the art. The nomenclatures used in connection with analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well-known and commonly used in the art unless otherwise indicated. [0090] So that the disclosure may be more readily understood, select terms are defined below.
DEFINITIONS
[0091] The terms "treat", "treating" and "treatment" refer to a method of alleviating or abrogating a disease and/or its attendant symptoms.
[0092] The term "subject" is defined herein to include animals such as mammals, including, but not limited to, primates (e.g., humans). In preferred embodiments, the subject is a human.
[0093] The terms“patient” and“subject” are used herein interchangeably.
[0094] The term“biological activity” refers to any one or more biological properties of a molecule (whether present naturally as found in vivo or provided or enabled by recombinant means). Biological properties include, but are not limited to, inhibiting tumor angiogenesis, inhibiting tumor-initiating/cancer stem cell maintenance, and inhibiting tumor cell chemoresi stance.
[0095] "Effective amount" refers to the amount sufficient to induce a desired biological, pharmacological, or therapeutic outcome in a subject. A therapeutically effective amount means a sufficient amount of a TRAIL receptor agonist protein in combination with anti-cancer agents to treat or prevent multiple myeloma, lung cancer, non-small cell lung cancer, colorectal cancer, and gastric cancer at a reasonable benefit/risk ratio applicable to any medical treatment. In addition, a therapeutically effective amount also means a sufficient amount of TRAIL receptor agonist protein in combination with anti-cancer agents to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone. It will be understood, however, that the usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment
[0096] “Specificity” refers to the ability of a binding protein to selectively bind an antigen or receptor.
[0097] The term“potency” refers to the ability of a binding protein to achieve a desired effect and is a measurement of its therapeutic efficacy. Potency may be assessed using methods known to one skilled in the art. [0098] The term“biological function” refers the specific in vitro or in vivo actions of a binding protein. Binding proteins may target several classes of antigens and achieve desired therapeutic outcomes through multiple mechanisms of action. Binding proteins may target soluble proteins, cell surface antigens, and/or extracellular protein deposits. Binding proteins may agonize, antagonize, or neutralize the activity of their targets. Binding proteins may assist in the clearance of the targets to which they bind or may result in cytotoxicity when bound to cells. Portions of two or more antibodies may be incorporated into a multivalent format to achieve more than one distinct function in a single binding protein molecule. In vitro assays and in vivo models used to assess biological function are known to one skilled in the art.
[0099] A“stable” binding protein is one in which the binding protein essentially retains its physical stability, chemical stability and/or biological activity upon storage. A multivalent binding protein that is stable in vitro at various temperatures for an extended period of time is desirable. Methods of stabilizing binding proteins and assessing their stability at various temperatures are known to one skilled in the art.
[00100] The term“solubility” refers to the ability of a protein to remain dispersed within an aqueous solution. The solubility of a protein in an aqueous formulation depends upon the proper distribution of hydrophobic and hydrophilic amino acid residues, and therefore, solubility can correlate with the production of correctly folded proteins. A person skilled in the art will be able to detect an increase or decrease in solubility of a binding protein using routine HPLC techniques and methods known to one skilled in the art.
[00101] “Control” refers to a composition that does not comprise an analyte (“negative control”) or does comprise the analyte (“positive control”). A positive control can comprise a known concentration of analyte. “Control,” “positive control,” and “calibrator” may be used interchangeably herein to refer to a composition comprising a known concentration of analyte. A “positive control” can be used to establish assay performance characteristics and is a useful indicator of the integrity of reagents (e.g., analytes).
[00102] The term“Fc region” defines the C-terminal region of an immunoglobulin heavy chain, which may be generated by papain digestion of an intact antibody. The Fc region may be a native sequence Fc region or a variant Fc region. The Fc region of an immunoglobulin generally comprises two constant domains, a CH2 domain and a CH3 domain, and optionally comprises a CH4 domain. Replacements of amino acid residues in the Fc portion to alter antibody effector function are known in the art (e.g., US Patent Nos. 5,648,260 and 5,624,821). The Fc region mediates several important effector functions, e.g., cytokine induction, antibody dependent cell mediated cytotoxicity (ADCC), phagocytosis, complement dependent cytotoxicity (CDC), and the half-life/clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for a therapeutic immunoglobulin but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives.
[00103] The term“linker” means an amino acid residue or a polypeptide comprising two or more amino acid residues joined by peptide bonds that are used to link two polypeptides. Examples of such linker polypeptides are well known in the art (see, e.g., Holliger et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak et al. (1994) Structure 2: 1121-1123).
[00104] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[00105] The use of the terms“a” and“an” and“the” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms“comprising,”“having,”“including,” and“containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g.,“such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non- claimed element as essential to the practice of the invention.
[00106] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
Table 1 provides the amino acid sequence for a receptor agonist protein monomer directed against TRAIL in ABBV-621.
Figure imgf000029_0001
Table 2 provides the full-length heavy and full-length light chain sequences for binding proteins directed against vascular endothelial growth factor (VEGF) in bevacizumab.
Figure imgf000030_0001
BRIEF DESCRIPTION OF THE SEQUENCE LISTING
[00107] Incorporated herein by reference in its entirety is a Sequence Listing entitled, “ABV12442WO01 ST25”, comprising SEQ ID NO: 1 through SEQ ID NO: 3, which includes the amino acid sequence disclosed herein. The Sequence listing has been submitted herewith in ASCII text format via EFS. The Sequence Listing was first created on March 11, 2019 and is 13 KB in size. EXAMPLES
EXAMPLE 1 - GROWTH INHIBITION OF XENOGRAFTED HUMAN COLORECTAL ADENOCARCINOMA TUMORS (DLD-1) BY ABBV-621 IN COMBINATION WITH
IRINOTECAN
Materials and Methods
Cell Culture Conditions:
[00108] DLD- 1 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). OPM-2 were obtained from the Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures (DSMZ, Germany). DLD-l and OPM-2 cells were maintained in RPMI 1640 medium (Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum.
Mice and Husbandry
[00109] Female SCID (DLD-l) and SCID Beige (OPM-2) mice were obtained from Charles River (Wilmington, MA). The body weight upon arrival was 18-20 g. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a l2-hour light: l2-hour dark schedule (lights on at 06:00 hours). All experiments were conducted in compliance with Abb Vie’ s Institutional Animal Care and Use Committee and the National Institutes of Health Guide for Care and Use of Laboratory Animals guidelines in a facility accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.
Parameters of Efficacy
[00110] Parameters of amplitude (tumor growth inhibition, TGI) and durability (tumor growth delay, TGD) of therapeutic response are used to refer to the efficacy of the drug. TGI indicates the divergence between the mean tumor volume of a drug-treated group and the mean tumor volume of the control and is expressed as a percentage of the mean volume of the control group. The TGI- value is determined at the last time point before the first animal is removed from a group because it reached its tumor burden limit. The TGD indicates the difference of the median time of a drug treated group to reach a defined tumor volume (mm3) as compared to the median time of a control group treated with vehicle to reach the same volume. This difference is expressed as a percentage of the median time of the control group to reach the specified tumor volume.
Compounds and Formulations
[00111] ABBV-621 is a TRAIL receptor agonist comprised of a human immunoglobulin Gl (IgGl)-Fc fused to native single chain TRAIL receptor binding domain (RBD) monomers that are covalently connected by glycosylated linkers, resulting in a dimer comprised of two sets of trimeric RBDs. The stock of ABBV-621 was maintained in a solution of 20 mM Tris, 70 g/L sucrose, 1.0 g/L polysorbate 80, pH 7.2 and stored at -80°C. Appropriate amounts of the stock were diluted in phosphate buffered saline prior to administration. Irinotecan was obtained from Teva (Irvine, CA), and formulated in phosphate buffered saline pH7.2. Bortezomib was obtained from Millenium Pharmaceuticals (Cambridge, MA), and formulated in phosphate buffered saline pH7.2.
Statistics
[00112] Statistically significant differences in mean tumor volume of compound-treated versus vehicle treated were assessed using the Student's t-test. Statistical analysis of survival was done using Log-Rank and Wilcoxon comparison with JMP software (SAS).
Generation of Tumor Bearing Mice and Determination of Tumor Volume of Subcutaneous Flank Tumors
[00113] For each subcutaneous study, viable cells were inoculated subcutaneously into the right flank of the mice on Day 0. The injection volume was 0.1 mL composed of a 1 : 1 mixture of S-MEM and Matrigel (BD, Franklin Lakes, NJ). All tumors were size matched at approximately 200-250 mm3 unless otherwise indicated. Therapy began within 24 hours after size matching the tumors. Mice weighed approximately 22 grams at the onset of therapy. Tumor volume was estimated two to three times weekly. Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the volume was calculated according to the following equation: V = L x W2/2. Mice were euthanized when tumor volume reached up to 3,000 mm3 or skin ulcerations occurred.
Experimental Conditions [00114] DLD-lcells were grown to passage three in vitro. Two million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 10. The mean tumor volume at tumor staging was approximately 206 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration were are described in Table 3.
Results and Discussion
[00115] Efficacy of ABBV-621 was determined in combination with Irinotecan. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was efficacious with TGI of 80% (p<0.00l). Irinotecan administered as monotherapy at 50 mg/kg Q4Dx4 was efficacious with TGI of 45% (p<0.05). As shown in Figure 1 / Table 3, the combination of ABBV-621 with Irinotecan was slightly greater than additive in terms of tumor growth inhibition as measured at the 42-day time period post-dose administration, and only slightly greater than additive for tumor growth delay using 1000 mm3 as the measured endpoint. However, this combination was determined to be synergistic in terms of the efficacy measures of overall response (OR), complete response (CR) and partial response (PR). All treatments groups were well tolerated, and no significant body weight loss was observed.
Table 3: Study Design to Determine Growth Inhibition of Xenografted Human Colorectal Adenocarcinoma Tumors (DLD-l) by ABBV- 621 in Combination with Irinotecan
Figure imgf000034_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 42 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response =
CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
EXAMPLE 2 - GROWTH INHIBITION OF XENOGRAFTED HUMAN MULTIPLE MYELOMA TUMORS (OPM-2) BY ABBV-621 IN COMBINATION WITH
BORTEZOMIB
Experimental Conditions.
[00116] OPM-2 cells were grown to passage three in vitro. One million cells per mouse were inoculated subcutaneously into the right flank of female SCID Beige mice on Day 0. Tumors were size matched on Day 22. The mean tumor volume at tumor staging was approximately 337 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. All other conditions were as described in Example 1. Details of the treatments and route of administration are described in Table 4.
Results and Discussion
[00117] Efficacy of ABBV-621 was determined in combination with Bortezomib. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was not efficacious with TGI of 34% (p>0.05). Bortezomib administered as monotherapy at 1 mg/kg Q4Dx3 was efficacious with TGI of 95% (p<0.00l). As shown in Figure 2 / Table 4, the combination of ABBV-621 and Bortezomib was only slightly additive in terms of tumor growth inhibition at measured at the 34-day time period post-dose administration but showed significant synergy in terms of tumor growth delay using 1000 mm3 as the measured endpoint. However, this combination was synergistic in terms of the efficacy measures of overall response (OR), complete response (CR) and partial response (PR). One mouse was found dead on day 37, while the rest of the treatment groups were well tolerated and with no significant body weight loss was observed. Table 4 - Study Design to Determine Growth Inhibition of Xenografted Human Multiple Myeloma Tumors (OPM-2) by ABBV-621 in Combination with Bortezomib.
Figure imgf000036_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 34 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response =
CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
EXAMPLE 3 - GROWTH INHIBITION OF XENOGRAFTED HUMAN NON-SMALL CELL LUNG CARCINOMA TUMORS (H460-LM) BY ABBV-621 IN COMBINATION
WITH PACLITAXEL
Materials and Methods
Cells and Culturing Conditions
[00118] NCI-H460LM was derived as a subclone of NCI-H460 at Abbvie from a lung metastasis tumor, and was maintained in RPMI 1640 medium (Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum.
Mice and Husbandry
[00119] Female SCID and SCID Beige mice were obtained from Charles River (Wilmington, MA). The body weight upon arrival was 18-20 g. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a l2-hour light: l2-hour dark schedule (lights on at 06:00 hours). All experiments were conducted in compliance with AbbVie’s Institutional Animal Care and Use Committee and the National Institutes of Health Guide for Care and Use of Laboratory Animals guidelines in a facility accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.
Parameters of Efficacy
[00120] Parameters of amplitude (tumor growth inhibition, TGI) and durability (tumor growth delay, TGD) of therapeutic response are used to refer to the efficacy of the drug. TGI indicates the divergence between the mean tumor volume of a drug-treated group and the mean tumor volume of the control and is expressed as a percentage of the mean volume of the control group. The TGI-value is determined at the last time point before the first animal is removed from a group because it reached its tumor burden limit. The TGD indicates the difference of the median time of a drug treated group to reach a defined tumor volume (mm3) as compared to the median time of a control group treated with vehicle to reach the same volume. This difference is expressed as a percentage of the median time of the control group to reach the specified tumor volume.
Compounds and Formulations [00121] ABBV-621 stock was maintained in a solution of 20 mM Tris, 70 g/L sucrose, 1.0 g/L polysorbate 80, pH 7.2 and stored at -80°C. Appropriate amounts of the stock were diluted in phosphate buffered saline prior to administration. Irinotecan was obtained from Hospira (Lake Forest, IL), and formulated in phosphate buffered saline. Paclitaxel was obtained from Clinigen Clinical Trial Services (Yardley, PA) and formulated in phosphate buffered saline.
Statistics
[00122] Statistically significant differences in mean tumor volume of compound-treated versus vehicle treated were assessed using the Student's t-test. Statistical analysis of survival was done using Log-Rank and Wilcoxon comparison with JMP software (SAS).
Generation of Tumor Bearing Mice and Determination of Tumor Volume of Subcutaneous Flank Tumors
[00123] For each subcutaneous study, viable cells were inoculated subcutaneously into the right flank of the mice on Day 0. The injection volume was 0.1 mL composed of a 1 :1 mixture of S- MEM and Matrigel (BD, Franklin Lakes, NJ). All tumors were size matched at approximately 200-250 mm3 unless otherwise indicated. Therapy began within 24 hours after size matching the tumors. Mice weighed approximately 22 grams at the onset of therapy. Tumor volume was estimated two to three times weekly. Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the volume was calculated according to the following equation: V = L x W2/2. Mice were euthanized when tumor volume reached up to 3,000 mm3 or skin ulcerations occurred.
Experimental Conditions.
[00124] H460-LM cells were grown to passage three in vitro. One million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 12. The mean tumor volume at tumor staging was approximately 300 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 5. Table 5 - Study Design to Determine Growth Inhibition of Xenografted Human Non-Small Cell Lung Carcinoma Tumors (H460-LM) by ABBV-621 in Combination with Paclitaxel
Figure imgf000039_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 20 measurements.
% TGD (tumor growth delay) = (T - C)/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
Results and Discussion
[00125] Efficacy of ABBV-621 was determined in combination with paclitaxel, a standard of care agent in non-small cell lung cancer. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx5 was efficacious with TGI of 31% (p<0.05). Paclitaxel administered as monotherapy at 30 mg/kg Q4Dx3 was not efficacious with TGI of 16%. As shown in Figure 3 / Table 5, the combination of ABBV-621 with Paclitaxel was synergistic in terms of tumor growth inhibition at the 20-day time period post-dose administration measurement. In addition, this combination was also synergistic in terms of the efficacy measures of overall response (OR) and partial response (PR). Two mice were found dead in the paclitaxel monotherapy group with no significant body weight loss. All other treatments groups were well tolerated, and no significant body weight loss was observed
EXAMPLE 4 - GROWTH INHIBITION OF XENOGRAFTED HUMAN COLORECTAL CARCINOMA TUMORS (SW-48) BY ABBV-621 IN COMBINATION WITH
IRINOTECAN
[00126] SW-48 were obtained from the American Type Culture Collection (ATCC,
Manassas, VA), and was maintained in DMEM medium (Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum (Hy clone, Logan, UT). All other conditions were as described in Example 3.
Experimental Conditions.
[00127] SW-48 cells were grown to passage three in vitro. Five million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 9. The mean tumor volume at tumor staging was approximately 251 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 6.
Results and Discussion
[00128] Efficacy of ABBV-621 was determined in combination with irinotecan, a standard of care agent in colorectal carcinoma. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx5 was efficacious with TGI of 39% (p<0.00l). Irinotecan administered as monotherapy at 50 mg/kg Q4Dx3 was efficacious with TGI of 73% (p<0.00l). As shown in Figure 4 / Table 6, the combination of ABBV-621 with Irinotecan was synergistic in terms of tumor growth tumor growth delay using 1000 mm3 as the measured endpoint. In addition, this combination was synergistic in terms of the efficacy measures of overall response, complete response and partial response. The irinotecan monotherapy group had significant body weight loss of 16%. All other treatments groups were well tolerated, and no significant body weight loss was observed.
Table 6 - Study Design to Determine Growth Inhibition of Xenografted Human Colorectal Carcinoma Tumors (SW-48) by ABBV- 621 in Combination with Irinotecan.
O
O
00
00
Figure imgf000042_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by
asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 24 measurements.
% TGD (tumor growth delay) = (T - C)/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint
of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle
control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population
n with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = H CR + PR. bno o o b4
*** p < 0.001, ** p < 0.005, * p<0.05 Ό 00
EXAMPLE 5 - GROWTH INHIBITION OF XENOGRAFTED HUMAN GASTRIC CARCINOMA TUMORS (Hs746t) BY ABBV-621 IN COMBINATION WITH
IRINOTECAN
Materials and Methods
Cell Culture Conditions:
[00129] Hs746t and NCI-N87 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). Hs746t cells were maintained in DMEM and NCI-N87 cells were in RPMI 1640 medium (Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum.
Mice and Husbandry
[00130] Female SCID mice were obtained from Charles River (Wilmington, MA). The body weight upon arrival was 18-20 g. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a l2-hour light: l2-hour dark schedule (lights on at 06:00 hours). All experiments were conducted in compliance with Abb Vie’ s Institutional Animal Care and Use Committee and the National Institutes of Health Guide for Care and Use of Laboratory Animals guidelines in a facility accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.
Parameters of Efficacy
[00131] Parameters of amplitude (tumor growth inhibition, TGI) and durability (tumor growth delay, TGD) of therapeutic response are used to refer to the efficacy of the drug. TGI indicates the divergence between the mean tumor volume of a drug-treated group and the mean tumor volume of the control and is expressed as a percentage of the mean volume of the control group. The TGI- value is determined at the last time point before the first animal is removed from a group because it reached its tumor burden limit. The TGD indicates the difference of the median time of a drug treated group to reach a defined tumor volume (mm3) as compared to the median time of a control group treated with vehicle to reach the same volume. This difference is expressed as a percentage of the median time of the control group to reach the specified tumor volume.
Compounds and Formulations [00132] ABBV-621 is a TRAIL receptor agonist comprised of a human immunoglobulin Gl (IgGl)-Fc fused to native single chain TRAIL receptor binding domain (RBD) monomers that are covalently connected by glycosylated linkers, resulting in a dimer comprised of two sets of trimeric RBDs. The stock of ABBV-621 was maintained in a solution of 20 mM Tris, 70 g/L sucrose, 1.0 g/L polysorbate 80, pH 7.2 and stored at -80°C. Appropriate amounts of the stock were diluted in phosphate buffered saline prior to administration. Irinotecan was obtained from Teva (Irvine, CA), and formulated in phosphate buffered saline pH7.2.
Statistics
[00133] Statistically significant differences in mean tumor volume of compound-treated versus vehicle treated were assessed using the Student's t-test. Statistical analysis of survival was done using Log-Rank and Wilcoxon comparison with JMP software (SAS).
Generation of Tumor Bearing Mice and Determination of Tumor Volume of Subcutaneous Flank Tumors
[00134] For each subcutaneous study, viable cells were inoculated subcutaneously into the right flank of the mice on Day 0. The injection volume was 0.1 mL composed of a 1 : 1 mixture of S-MEM and Matrigel (BD, Franklin Lakes, NJ). All tumors were size matched at approximately 200-250 mm3 unless otherwise indicated. Therapy began within 24 hours after size matching the tumors. Mice weighed approximately 22 grams at the onset of therapy. Tumor volume was estimated two to three times weekly. Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the volume was calculated according to the following equation: V = L x W2/2. Mice were euthanized when tumor volume reached up to 3,000 mm3 or skin ulcerations occurred.
Experimental Conditions
[00135] Hs746t cells were grown to passage three in vitro. Two million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 13. The mean tumor volume at tumor staging was approximately 257 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 7. Results and Discussion
[00136] Efficacy of ABBV-621 was determined in combination with Irinotecan in the Hs746t model. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was efficacious with TGI of 41% (p<0.00l). Irinotecan administered as monotherapy at 50 mg/kg Q4Dx3 was efficacious with TGI of 94% (p<0.00l). As shown in Figure 5 / Table 7, the combination of ABBV-621 with Irinotecan showed increased efficacy when compared to irinotecan alone both by TGI (99.5%; p<0.00l) and by tumor growth delay (218%; p<0.005) using 1000 mm3 as the measured endpoint. Tumors treated with the combination also resulted in a significant increase in complete responses. All treatments groups were well tolerated, and no significant body weight loss was observed.
Table 7: Study Design to Determine Growth Inhibition of Xenografted Human Gastric carcinoma Tumors (Hs746t) by ABBV-621 in Combination with Irinotecan
Figure imgf000046_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 27 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
EXAMPLE 6 - GROWTH INHIBITION OF XENOGRAFTED HUMAN GASTRIC CARCINOMA TUMORS (Hs746t) BY ABBV-621 IN COMBINATION WITH
DOCETAXEL
Experimental Conditions
[00137] Hs746t cells were grown to passage three in vitro. Two million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 13. The mean tumor volume at tumor staging was approximately 257 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 8. All other conditions were as described in Example 5.
Results and Discussion
[00138] Efficacy of ABBV-621 was determined in combination with docetaxel in the Hs746t model. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was efficacious with TGI of 41% (p<0.00l). Docetaxel administered as monotherapy at 5 mg/kg QDxl was efficacious with TGI of 90% (p<0.00l). As shown in Figure 6 / Table 8, the combination of ABBV-621 with docetaxel showed increased efficacy when compared to docetaxel alone both by TGI (87%; p<0.00l) and by tumor growth delay (13%; p<0.005) using 1000 mm3 as the measured endpoint. Tumors treated with the combination also resulted in a significant increase in partial, complete, and overall responses. All treatments groups were well tolerated, and no significant body weight loss was observed.
Table 8: Study Design to Determine Growth Inhibition of Xenografted Human Gastric carcinoma Tumors (Hs746t) by ABBV-621 in Combination with Docetaxel
Figure imgf000048_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 27 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
EXAMPLE 7 - GROWTH INHIBITION OF XENOGRAFTED HUMAN GASTRIC CARCINOMA TUMORS (NCI-N87) BY ABBV-621 IN COMBINATION WITH
IRINOTECAN
Experimental Conditions
[00139] NCI-N87 cells were grown to passage three in vitro. Two million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 13. The mean tumor volume at tumor staging was approximately 252 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 9. All other conditions were as described in Example 5.
Results and Discussion
[00140] Efficacy of ABBV-621 was determined in combination with Irinotecan in the NCI-N87 model. Neither ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 nor irinotecan administered as monotherapy at 50 mg/kg Q4Dx3 showed significant efficacy alone. As shown in Figure 7 / Table 9, the combination of ABBV-621 with Irinotecan showed increased efficacy when compared to irinotecan alone both by TGI (60%; p<0.00l) and by tumor growth delay (58%; p<0.005) using 1000 mm3 as the measured endpoint. Tumors treated with the combination also resulted in a significant increase in partial and overall responses. All treatments groups were well tolerated and no significant body weight loss was observed.
Table 9: Study Design to Determine Growth Inhibition of Xenografted Human Gastric carcinoma Tumors (NCI-N87) by ABBV-621 in
Combination with Irinotecan
O
O
00
00
Figure imgf000050_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by
asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 83 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint
of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle
control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population
n with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = H CR + PR. n
O
O
* * * p < 0.001, * * p < 0.005, * p<0.05
Ό
00
EXAMPLE 8 - GROWTH INHIBITION OF XENOGRAFTED HUMAN GASTRIC CARCINOMA TUMORS (NCI-N87) BY ABBV-621 IN COMBINATION WITH
DOCETAXEL
Experimental Conditions
[00141] NCI-N87 cells were grown to passage three in vitro. Two million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 13. The mean tumor volume at tumor staging was approximately 252 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 10. All other conditions were as described in Example 5.
Results and Discussion
[00142] Efficacy of ABBV-621 was determined in combination with docetaxel in the NCI-N87 model. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was not efficacious in this model. Docetaxel administered as monotherapy at 5 mg/kg QDxl was efficacious with TGI of 55% (p<0.00l). As shown in Figure 8 / Table 10, the combination of ABBV-621 with docetaxel showed increased efficacy when compared to docetaxel alone both by TGI (85%; p<0.00l) and by tumor growth delay (63%; p<0.005) using 1000 mm3 as the measured endpoint. Tumors treated with the combination also resulted in significant increases in partial and overall responses. All treatments groups were well tolerated and no significant body weight loss was observed.
Table 10: Study Design to Determine Growth Inhibition of Xenografted Human Gastric carcinoma Tumors (NCI-N87) by ABBV-621 in Combination with Docetaxel
Figure imgf000052_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 83 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response =
CR + PR.
*** p < 0.001, ** p < 0.005, * p<0.05
EXAMPLE 10 - STRONG RESPONSES TO ABBV-621 ARE ENRICHED WITH ACTIVATING KRAS MUTATIONS IN COLORECTAL CARCINOMA TUMOR PDX
MODELS
Materials and Methods
[00143] Efficacy of ABBV-621 was determined in colorectal carcinoma patient derived xenograft models. 23 CRC PDx models mice were treated with ABBV-621 (3 mg/kg/day, Q2Dx5, IP. For each model, vehicle treated (n = 1) and ABBV-621 treated (n = 3) tumor bearing mice were evaluated for 60 days post initiation of treatment.). The KRAS status for the late passage PDx tumors (passage used for the testing of efficacy of ABBV-621) were identified by whole exome sequencing and were confirmed by the Qiagen’ s Somatic mutation PCR kit. mRNA levels for DR4 and 5 were estimated by full transcriptome analysis using RNAseq.
[00144] Quantitative PCR (qPCR): Real-time PCR was performed on DNA extracted from formalin-fixed paraffin-embedded (FFPE) tumor samples collected from PDX models.
qBiomarker™ Somatic Mutation PCR Array Human KRAS Gene Kit from Qiagen was used to perform this testing (Product No.: 337021, Cat. No.: SMH-806AA). 20 hot-spot KRAS mutations were included on the panel (Table 11). Allele-specific amplification was achieved by Amplification Refractory Mutation System (ARMS) technology, which is based on the discrimination by Taq polymerase between a match and a mismatch at the 3’ end of the PCR primer. DNA template was mixed with ready -to-use PCR mastermix, equal volumes were aliquoted to each well of the same plate, and then run on the real-time PCR cycling program. (Bio-Rad CFX96). Data was analyzed using the available Excel-based data analysis templates provided by Qiagen (copyright protected).
[00145] Whole Exome Sequencing (WES): Genomic DNA was sonicated to an average size of approximately 200 bp. The fragments were blunt ended, A’ tailed and ligated to Illumina sequencing adapters. The ligated fragments were amplified for 7 PCR cycles. Exome targets were enriched with custom Agilent SureSelect baits according to the manufacturer’s protocols. Enriched fragments were amplified for 14 cycles with primers that incorporate a unique indexing sequence tag. The resulting library fragments were sequenced from each end (paired-end reads) for 150 bases using an Illumina HiSeq-3000 instrument.
[00146] RNAseq: Total RNA integrity was determined using Agilent Bioanalyzer or 4200 Tapestation. Library preparation was performed with 1-50 ng of total RNA. ds-cDNA was prepared using the SeqPlex RNA Amplification Kit (Sigma) per manufacturer’s protocol. cDNA was blunt ended, had an A base added to the 3’ ends, and then had Illumina sequencing adapters ligated to the ends. Ligated fragments were then amplified for 12 cycles using primers incorporating unique index tags. Fragments were sequenced on an HiSeq-3000 using single reads extending 50 bases. DR4 and DR5 mRNA levels in KRAS mutant (gain of function) or wild type (WT) colorectal PDx tumors is illustrated in Figure 9. DR4 and DR5 mRNA levels in KRAS mutant (gain of function) or wild type (WT) colorectal PDx tumors is illustrated in Figure 9. DR4 and DR5 mRNA levels in KRAS mutant (gain of function) or wild type (WT) PDx tumors (various indications) is illustrated in Figure 10.
Table 11 : Summary of Growth Inhibition of Patient Derived Xenografted Human Colorectal Carcinoma Tumors by ABBV-621. PDX models were treated with ABBV-621 at 3 mg/kg/day, Q2Dx5, IP. For each model, vehicle treated (n = 1) and ABBV-621 treated (n = 3) tumor bearing mice were evaluated for 60 days post initiation of treatment. Efficacy results were grouped into High (initial efficacy that is maintained during study), Low (initial efficacy with regrowth), or No (no efficacy).
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Results and Conclusions
[00147] 13/23 CRC PDXs had KRAS gain of function mutations. CRC PDXs with KRAS mutation had higher response to single agent ABBV-621 compared with PDXs with wild type KRAS. Specifically 12/15 (80%) High/Strong responses were in PDXs with KRAS mutations. Only 1/8 (12.5%) Moderate/Negative responses possess KRAS mutations. DR4 and DR5 mRNA levels are higher in KRAS mutant (gain of function) tumors compared to KRAS WT tumors, and higher levels of death receptors may contribute to the increased sensitivity to ABBV- 621.
EXAMPLE 11 - GROWTH INHIBITION OF XENOGRAFTED HUMAN PATIENT DERIVED COLORECTAL CARCINOMA TUMORS BY ABBV-621
Materials and Methods
Cell Culture Conditions: [00148] SW-48, Colo-205, and DLD-l cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). SW-48 cells were maintained in DMEM (Invitrogen, Carlsbad, CA) and supplemented with 10% fetal bovine serum. Colo-205 and DLD-l cells were maintained in RPMI (Invitrogen, Carlsbad, CA) and supplemented with 10% fetal bovine serum.
Mice and Husbandry
[00149] Female SCID, female SCID beige, and female nude mice were obtained from Charles River (Wilmington, MA). For the colorectal PDX studies performed at Champions Oncology, female nude mice were obtained from Harlan Laboratories (Indianapolis, IN). The body weight upon arrival was 18-20 g. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a l2-hour light: l2-hour dark schedule (lights on at 06:00 hours). All experiments were conducted in compliance with Abb Vie’ s Institutional Animal Care and Use Committee and the National Institutes of Health Guide for Care and Use of Laboratory Animals guidelines in a facility accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.
Parameters of Efficacy
[00150] Parameters of amplitude (tumor growth inhibition, TGI) and durability (tumor growth delay, TGD) of therapeutic response are used to refer to the efficacy of the drug. TGI indicates the divergence between the mean tumor volume of a drug-treated group and the mean tumor volume of the control and is expressed as a percentage of the mean volume of the control group. The TGI- value is determined at the last time point before the first animal is removed from a group because it reached its tumor burden limit. The TGD indicates the difference of the median time of a drug treated group to reach a defined tumor volume (mm3) as compared to the median time of a control group treated with vehicle to reach the same volume. This difference is expressed as a percentage of the median time of the control group to reach the specified tumor volume. Compounds and Formulations
[00151] ABBV-621 is a TRAIL receptor agonist comprised of a human immunoglobulin Gl (IgGl)-Fc fused to native single chain TRAIL receptor binding domain (RBD) monomers that are covalently connected by glycosylated linkers, resulting in a dimer comprised of two sets of trimeric RBDs. The stock of ABBV-621 was maintained in a solution of 20 mM Tris, 70 g/L sucrose, 1.0 g/L polysorbate 80, pH 7.2 and stored at -80°C. Appropriate amounts of the stock were diluted in phosphate buffered saline prior to administration. Irinotecan was obtained from Teva (Irvine, CA), and formulated in phosphate buffered saline pH7.2.
Statistics
[00152] Statistically significant differences in mean tumor volume of compound-treated versus vehicle treated were assessed using the Student's t-test. Statistical analysis of survival was done using Log-Rank and Wilcoxon comparison with JMP software (SAS).
Generation of Tumor Bearing Mice and Determination of Tumor Volume of Subcutaneous Flank Tumors
[00153] For the cell line based subcutaneous studies (Colo-205, DLD-l, and SW48), viable cells were inoculated subcutaneously into the right flank of the mice on Day 0. The injection volume was 0.1 mL composed of a 1 : 1 mixture of S-MEM and Matrigel (BD, Franklin Lakes, NJ). For the patient derived xenograft subcutaneous studies (CTG-0064 and CTG-0069), fresh tumor tissue was cut into fragments and implanted. All tumors were size matched at approximately 200-250 mm3 unless otherwise indicated. Therapy began within 24 hours after size matching the tumors. Mice weighed approximately 22 grams at the onset of therapy. Tumor volume was estimated two to three times weekly. Measurements of the length (L) and width (W) of the tumor were taken via electronic calliper and the volume was calculated according to the following equation: V = L x W2/2. Mice were euthanized when tumor volume reached up to 3,000 mm3 or skin ulcerations occurred. Experimental Conditions
[00154] Colo-205 cells were grown to passage 3 in vitro. 2 million cells per mouse were inoculated subcutaneously into the right flank of female SCID beige mice on Day 0. Tumors were size matched on Day 9. The mean tumor volume at tumor staging was approximately 247 mm3. Data calculations are made and stored using Study log, Study director
Version 3.1.268.184. Details of the treatments and route of administration are described in Table 12
[00155] DLD-l cells were grown to passage 3 in vitro. 2 million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 10. The mean tumor volume at tumor staging was approximately 206 mm3. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 3.
[00156] SW-48 cells were grown to passage 3 in vitro. 5 million cells per mouse were inoculated subcutaneously into the right flank of female SCID mice on Day 0. Tumors were size matched on Day 9. The mean tumor volume at tumor staging was approximately 251 mm3.
Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Table 13.
[00157] CTG-0064 or CTG-0069 tumor bearing mice were used to provide tumor tissue for propagation into study mice. Tumors were excised and cut into 2 mm x 2 mm fragments.
Fragments were placed subcutaneously using a trocar. Data calculations are made and stored using Study log, Study director Version 3.1.268.184. Details of the treatments and route of administration are described in Tables 14, 15.
Results and Discussion
[00158] Efficacy of ABBV-621 was determined in combination with irinotecan in the Colo-205 KRAS wild-type model. ABBV-621 administered as monotherapy at 0.3 mg/kg Q2Dx 5 was efficacious with TGI of 38% (p<0.05). Irinotecan administered as monotherapy at 30 mg/kg Q3Dx3 was efficacious with TGI of 59% (p<0.00l). As shown in Figure 11 / Table 12, the combination of ABBV-621 with Irinotecan showed increased efficacy when compared to irinotecan alone both by TGI (41%; p<0.0l) and by tumor growth delay (21%; p<0.05) using 1000 mm3 as the measured endpoint. All treatments groups except the irinotecan monotherapy group were well tolerated and no significant body weight loss was observed. In the irinotecan treated group, there was 16% weigh loss after the 3rd dose, while the combination group only had 11% weight loss.
Table 12: Study Design to Determine Growth Inhibition of Xenografted Human Colorectal Carcinoma Tumors (Colo-205) by ABBV- 621 in Combination with Irinotecan
o
O
00
00
Figure imgf000061_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as
indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day
24 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time
to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle control group. Based on an endpoint of 1000 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of n H population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements;
n OR: overall response = CR + PR. O
O
*** p < 0.001, ** p < 0.01, * p<0.05
0©0
[00159] Efficacy of ABBV-621 was determined in combination with irinotecan in the DLD-l KRAS mutant model. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was efficacious with TGI of 80% (p<0.00l). Irinotecan administered as monotherapy at 50 mg/kg Q4Dx4 was efficacious with TGI of 45% (p<0.00l 1). As shown in Figure 1 / Table 3, the combination of ABBV-621 with irinotecan showed increased efficacy when compared to irinotecan alone both by TGI (77%; p<0.005) and by tumor growth delay (>30%; p<0.0l) using 1000 mm3 as the measured endpoint. All treatments groups were well-tolerated and no significant body weight loss was observed.
[00160] Efficacy of ABBV-621 was determined in combination with irinotecan in the SW-48 KRas wild-type model. ABBV-621 administered as monotherapy at 3 mg/kg Q2Dx 5 was efficacious with TGI of 39% (p<0.00l). Irinotecan administered as monotherapy at 50 mg/kg Q4Dx3 was efficacious with TGI of 73% (p<0.00l). As shown in Figure 12 / Table 13, the combination of ABBV-621 with irinotecan showed increased efficacy when compared to irinotecan alone both by TGI (97%; p<0.00l) and by tumor growth delay (127%; p<0.00l) using 1000 mm3 as the measured endpoint. All treatments groups except Irinotecan group were well tolerated and no significant body weight loss was observed. In Irinotecan treated group, there was 16% weigh loss on day 24.
Table 13 : Study Design to Determine Growth Inhibition of Xenografted Human Colorectal Carcinoma Tumors (SW-48) by ABBV-621
in Combination with Irinotecan
O
O
00
00
Figure imgf000063_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as
indicated by asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day
24 measurements.
% TGD (tumor growth delay) = (T - C )/ C x 100, where T is the median time to endpoint of treatment group and C is the median time
to endpoint of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment
n group vs. vehicle control group. Based on an endpoint of 1000 mm3. H
n
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of O population with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; O OR: overall response = CR + PR. Ό
00
*** p < 0.001, ** p < 0.005, * p<0.05
[00161] Efficacy of ABBV-621 was determined in combination with irinotecan in the CTG- KRAS mutant 0064 model. ABBV-621 administered as monotherapy at 0.3 mg/kg Q2Dx 5 was efficacious with TGI of 47% (p<0.0l). Irinotecan administered as monotherapy at 25 mg/kg Q4Dx3 was efficacious with TGI of 68% (p<0.00l). As shown in Figure 13 / Table 14, while the combination of ABBV-621 with irinotecan showed a slight increase in efficacy when compared to irinotecan alone, a significant difference was not observed by either TGI (47%; NS) or tumor growth delay (53%; NS) using 1000 mm3 as the measured endpoint. All treatments groups were well tolerated and no significant body weight loss was observed.
Table 14: Study Design to Determine Growth Inhibition of Patient Derived Xenografted Human Colorectal Carcinoma Tumors (CTG- 0064) by ABBV-621 in Combination with Irinotecan
O
O
00
00
Figure imgf000065_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by
asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 52 measurements.
% TGD (tumor growth delay) = (T - C)/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint
of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle
control group. Based on an endpoint of 1000 mm3. n H
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population bn
O
with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response =
O b
CR + PR.
so
*** p < 0.001, ** p < 0.005, * p<0.05 00
[00162] Efficacy of ABBV-621 was determined in combination with docetaxel in the CTG-0069 KRAS mutant model. ABBV-621 administered as monotherapy at 1 mg/kg Q2Dx 5 was efficacious with TGI of 96% (p<0.00l). Irinotecan administered as monotherapy at 12.5 mg/kg Q4Dx3 was efficacious with TGI of 91% (p<0.00l). As shown in Figure 14 / Table 15, the combination of ABBV-621 with irinotecan showed increased efficacy when compared to ABBV-621 alone both by TGI (87%; p<0.00l) and by tumor growth delay (61%; p<0.05) using 400 mm3 as the measured endpoint. All treatments groups were well tolerated and no significant body weight loss was observed.
Table 15: Study Design to Determine Growth Inhibition of Patient Derived Xenografted Human Colorectal Carcinoma Tumors (CTG- 0069) by ABBV-621 in Combination with Irinotecan
O
O
00
00
Figure imgf000067_0001
%TGI (tumor growth inhibition) = 1 - (mean tumor volume of treatment group/ tumor volume of vehicle control group). P values (as indicated by
asterisks) are derived from Student’s T test comparison of treatment group vs. treatment control group (a). Based on day 36 measurements.
% TGD (tumor growth delay) = (T - C)/ C x 100, where T is the median time to endpoint of treatment group and C is the median time to endpoint
of vehicle control group. P values (as indicated by asterisks) derived from Kaplan Meier log-rank comparison of treatment group vs. vehicle
control group. Based on an endpoint of 400 mm3.
CR: complete response, % of population with tumors < 25 mm3 for at least 3 consecutive measurements; PR: partial response, % of population n H with tumors > 25 mm3 and < 50% of the original tumor volume at onset of dosing for at least 3 consecutive measurements; OR: overall response = n
O
CR + PR.
O
*** p < 0.001, ** p < 0.005, * p<0.05 b4 s 0o0

Claims

WE CLAIM:
1. A method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with one or more anti-cancer agents.
2. The method of Claim 1 , wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer.
3. The method of Claim 1, wherein the anti-cancer agent is selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, bortezomib, and docetaxel.
4. The method of Claim 3, wherein the anti-cancer agent is administered prior to, subsequent to, or concurrently with ABBV-621.
5. The method of Claim 3, wherein the anti-cancer agent is bortezomib.
6. The method of Claim 3, wherein the anti-cancer agent is paclitaxel.
7. The method of Claim 3, wherein the anti-cancer agent is irinotecan.
8. The method of Claim 3, wherein the anti-cancer agent is selected from the group consisting of: (i) FOLFIRI; and (ii) FOLFIRI and bevacizumab.
9. The method of Claim 3, wherein the anti-cancer agent is docetaxel.
10. A method for the treatment of cancer for a subject in need thereof, comprising administering to the subject a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV- 621, in combination with one or more anti-cancer agents selected from the group consisting of: paclitaxel, irinotecan, FOLFIRI, bortezomib, and docetaxel, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone.
11. The method of Claim 10, wherein the cancer is selected from the group consisting of: multiple myeloma, gastric cancer, lung cancer, non-small cell lung cancer, and colorectal cancer.
12. The method of Claim 10, wherein the anti-cancer agent is administered prior to, subsequent to, or concurrently with ABBV-621.
13. The method of Claim 10, wherein the anti-cancer agent is bortezomib.
14. The method of Claim 10, wherein the anti-cancer agent is paclitaxel.
15. The method of Claim 10, wherein the anti-cancer agent is irinotecan.
16. The method of Claim 10, wherein the anti-cancer agent is selected from the group consisting of: (i) FOLFIRI; and (ii) FOLFIRI and bevacizumab.
17. The method of Claim 10, wherein the anti-cancer agent is docetaxel.
18. A method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the TRAIL receptor agonist ABBV-621 or a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of: irinotecan, FOLFIRI, and bevacizumab, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative
to the administration of either agent alone.
19. A method for the treatment of colorectal KRAS mutant positive cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the TRAIL receptor agonist ABBV-621 or a synergistic, therapeutically effective amount of the TRAIL receptor agonist ABBV-621, in combination with one or more anti-cancer agents selected from the group consisting of: irinotecan, FOLFIRI, and bevacizumab, wherein the subject is able to achieve enhanced tumor inhibition, tumor growth delay, and/or enhanced efficacy relative to the administration of either agent alone; wherein said patient also has elevated expression of DR4 and/or DR5.
PCT/US2019/022398 2018-03-15 2019-03-15 Abbv-621 in combination with anti-cancer agents for the treatment of cancer WO2019178438A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862643242P 2018-03-15 2018-03-15
US62/643,242 2018-03-15
US201962815788P 2019-03-08 2019-03-08
US62/815,788 2019-03-08

Publications (1)

Publication Number Publication Date
WO2019178438A1 true WO2019178438A1 (en) 2019-09-19

Family

ID=65952176

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/022398 WO2019178438A1 (en) 2018-03-15 2019-03-15 Abbv-621 in combination with anti-cancer agents for the treatment of cancer

Country Status (2)

Country Link
TW (1) TW202003553A (en)
WO (1) WO2019178438A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4604463A (en) 1983-07-14 1986-08-05 Kabushiki Kaisha Yakult Honsha Camptothecin derivatives and process for preparing same
US4814470A (en) 1986-07-17 1989-03-21 Rhone-Poulenc Sante Taxol derivatives, their preparation and pharmaceutical compositions containing them
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5780454A (en) 1994-10-28 1998-07-14 Proscript, Inc. Boronic ester and acid compounds
US8980556B2 (en) 2011-01-11 2015-03-17 Roche Molecular Systems, Inc. High resolution melting analysis as a prescreening tool
WO2015164588A1 (en) * 2014-04-23 2015-10-29 Abbvie, Inc. Single-chain trail-receptor agonist proteins

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4604463A (en) 1983-07-14 1986-08-05 Kabushiki Kaisha Yakult Honsha Camptothecin derivatives and process for preparing same
US4814470A (en) 1986-07-17 1989-03-21 Rhone-Poulenc Sante Taxol derivatives, their preparation and pharmaceutical compositions containing them
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5780454A (en) 1994-10-28 1998-07-14 Proscript, Inc. Boronic ester and acid compounds
US8980556B2 (en) 2011-01-11 2015-03-17 Roche Molecular Systems, Inc. High resolution melting analysis as a prescreening tool
WO2015164588A1 (en) * 2014-04-23 2015-10-29 Abbvie, Inc. Single-chain trail-receptor agonist proteins
US20150337027A1 (en) 2014-04-23 2015-11-26 Abbvie Inc. Single-chain trail-receptor agonist proteins

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
ANDRE, T. ET AL., ANNALS OF ONCOLOGY, vol. 00, 2012, pages 1 - 8
ANONYMOUS: "Abbv-621 Is a Novel and Potent TRAIL Receptor Agonist Fusion Protein That Induces Apoptosis Alone and in Combination with Navitoclax and Venetoclax in Hematological Tumors | Blood Journal", 1 January 2017 (2017-01-01), XP055590747, Retrieved from the Internet <URL:http://www.bloodjournal.org/content/130/Suppl_1/2812?sso-checked=true> [retrieved on 20190521] *
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 100286-90-6
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 179324-69-7
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 33069-62-4
DEROOK, W. ET AL., LANCIT ONCOLOGY, vol. 11, 2010, pages 6753 - 6762
HOLLIGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
M. C. WANI ET AL., J. AM. CHEM. SOC., vol. 93, May 1971 (1971-05-01), pages 2325
POLJAK ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123
YUAN XUN ET AL: "Developing TRAIL/TRAIL death receptor-based cancer therapies", CANCER METASTASIS, KLUWER ACADEMIC PUBLISHERS, DORDRECHT, NL, vol. 37, no. 4, 14 March 2018 (2018-03-14), pages 733 - 748, XP036690678, ISSN: 0167-7659, [retrieved on 20180314], DOI: 10.1007/S10555-018-9728-Y *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder

Also Published As

Publication number Publication date
TW202003553A (en) 2020-01-16

Similar Documents

Publication Publication Date Title
Sun et al. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends
TWI795347B (en) Treatment of lung cancer using a combination of an anti-pd-1 antibody and an anti-ctla-4 antibody
KR20170074962A (en) Combination therapy for cancer
ES2742079T3 (en) Treatment of Waldenström macroglobulinemia associated with C1013G / CXCR4 with an anti-CXCR4 antibody
Punekar et al. Immunotherapy in non-small cell lung cancer: Past, present, and future directions
KR20230151057A (en) Use of 1-[4-bromo-5-[1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl]-2-fluorophenyl]-3-phenylurea and analogs for the treatment of cancers associated with genetic abnormalities in platelet derived growth factor receptor alpha
CN112218658A (en) Use of caloric restriction mimetics for enhancing chemoimmunotherapy for cancer treatment
WO2019178438A1 (en) Abbv-621 in combination with anti-cancer agents for the treatment of cancer
CA3143339A1 (en) Patient selection for enhancement of anti-tumor immunity in cancer patients
US20210188999A1 (en) Abcb5 ligands and substrates
CN116056710A (en) Treatment of cancer with TLR agonists
CN113939309A (en) Treatment of cancer with sEphB4-HSA fusion protein
AU2018314227A1 (en) Treatment of triple negative breast cancer or colorectal cancer with liver metastases with an anti PD-L1 antibody and an oncolytic virus
Daisy Precilla et al. Disentangling the therapeutic tactics in GBM: From bench to bedside and beyond
Li et al. Obatoclax, the pan-Bcl-2 inhibitor sensitizes hepatocellular carcinoma cells to promote the anti-tumor efficacy in combination with immune checkpoint blockade
TW202039555A (en) USE OF IL-1β BINDING ANTIBODIES
Shigetomi et al. Targeted molecular therapies for ovarian cancer: an update and future perspectives
US20220056123A1 (en) Use of il-1beta binding antibodies
CN111278448A (en) Compositions and methods for treating liver cancer
US20220096436A1 (en) Combination product for the treatment of cancer
US9566334B2 (en) Combinations of a PI3K/AKT inhibitor compound with an HER3/EGFR inhibitor compound and use thereof in the treatment of a hyperproliferative disorder
WO2019178433A1 (en) Abbv-621 in combination with anti-cancer agents for the treatment of pancreatic cancer
US20240156908A1 (en) USE OF sEphB4-HSA FUSION PROTEIN AS A FIRST-LINE THERAPY IN CANCER TREATMENT
JP2022549337A (en) Composite biomarkers for cancer therapy
WO2021009761A1 (en) Combination therapy for the treatment of cancer

Legal Events

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

Ref document number: 19714026

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19714026

Country of ref document: EP

Kind code of ref document: A1