Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ABBV-621 IN COMBINATION WITH ANTI-CANCER AGENTS FOR THE TREATMENT OF PANCREATIC CANCER
Document Type and Number:
WIPO Patent Application WO/2019/178433
Kind Code:
A1
Abstract:
This invention pertains to a method for the treatment of pancreatic cancer in a subject comprising administering to the subject an effective amount of ABBV-621 in combination with anti-cancer agents.

Inventors:
MORGAN-LAPPE SUSAN E (US)
PHILLIPS DARREN C (US)
SOLOMON LARRY R (US)
BUCHANAN FRITZ G (US)
Application Number:
PCT/US2019/022392
Publication Date:
September 19, 2019
Filing Date:
March 15, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ABBVIE INC (US)
International Classes:
A61K31/00; A61K31/337; A61K31/4745; A61K31/69; A61K33/00; A61P35/00; C07K14/195; C07K14/435; C07K14/44; C07K14/705
Domestic Patent References:
WO2015164588A12015-10-29
WO2010138588A22010-12-02
WO2013055897A12013-04-18
Foreign References:
US20150337027A12015-11-26
GB2136425A1984-09-19
US2802005A1957-08-06
USRE41065E2009-12-29
US4814470A1989-03-21
JPS6019790A1985-01-31
US7390799B22008-06-24
US5648260A1997-07-15
US5624821A1997-04-29
Other References:
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), XP055591461, Retrieved from the Internet [retrieved on 20190523]
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
M. MOSCOSO CASTRO ET AL: "American Association for Cancer Research (AACR) - 108th Annual Meeting. Washington, D.C., USA - April 1-5, 2017", DRUGS OF THE FUTURE, vol. 42, no. 6, 5 April 2017 (2017-04-05), ES, pages 359 - 366, XP055475574, ISSN: 0377-8282, DOI: 10.1358/dof.2017.042.06.2634466
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 51-28-8
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 183321-74-6
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 114977-28-5
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 97682-44-5
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 923564-51-6
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1257044-40-8
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1430845-59-2
HOLLIGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
POLJAK ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123
BORISY ET AL., PROC NATL ACAD SCI USA, vol. 100, 2003, pages 7977 - 7982
Attorney, Agent or Firm:
D'AMICO, Stephen C. et al. (US)
Download PDF:
Claims:
WE CLAIM:

1. A method for the treatment of pancreatic 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 one or more anti-pancreatic cancer agents.

2. The method of claim 1, wherein the anti-pancreatic cancer agent is selected from the group consisting of: gemcitabine, 5-fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan.

3. The method of claim 2, wherein the anti -pancreatic cancer agent is administered prior to, after, or concomitantly with ABBV-621.

4. The method of claim 2, wherein the anti-pancreatic cancer agent is gemcitabine.

5. The method of claim 2, wherein the anti -pancreatic cancer agents is 5-fluorouracil.

6. The method of claim 2, wherein the anti-pancreatic cancer agents is erlotinib.

7. The method of claim 2, wherein the anti-pancreatic cancer agents is paclitaxel.

8. The method of claim 2, wherein the anti -pancreatic cancer agents is irinotecan.

9. The method of claim 1, wherein ABBV-621 is dosed in combination with an apoptosis inducing anti-cancer agents selected from the group consisting of: navitoclax, venetoclax, and Compound (I).

10. The method of claim 9, wherein the apoptosis inducing anti-cancer agent is navitoclax.

11. The method of claim 9, wherein the apoptosis inducing anti-cancer agent is venetoclax.

12. The method of claim 9, wherein the apoptosis inducing anti-cancer agent is Compound (I).

13. The method of claim 2, further comprising the administration of nab -paclitaxel.

14. The method of claim 2, wherein said paclitaxel is nab -paclitaxel.

15. The method of claim 2, wherein the anti -pancreatic cancer agent is docetaxel

16. A method for the treatment of pancreatic 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 the anti-pancreatic cancer agent, gemcitabine.

17. A method for the treatment of pancreatic 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 the anti-pancreatic cancer agents, gemcitabine and nab- paclitaxel.

18. A method for the treatment of pancreatic 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 -pancreatic cancer agent selected from the group consisting of: paclitaxel and nab-paclitaxel.

19. A method for the treatment of pancreatic 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 the anti-pancreatic cancer agent, SN-38.

20. A method for the treatment of pancreatic 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 the anti-pancreatic cancer agent, docetaxel.

Description:
ABBV-621 IN COMBINATION WITH ANTI-CANCER AGENTS FOR THE

TREATMENT OF PANCREATIC CANCER

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application Serial No.

62/643,235, filed March 15, 2018; as well as claims the benefit of U.S. Provisional Application Serial No. 62/815,549, filed March 8, 2019, the disclosures of which are hereby incorporated by reference in their entirety.

FIELD OF THE INVENTION

[0001] This invention pertains to synergistic methods of using ABBV-621 in combination with anti-cancer agents for the treatment of pancreatic cancer.

BACKGROUND OF THE INVENTION

[0002] Cell death can be initiated through activation of the extrinsic and intrinsic apoptotic signaling 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 signaling 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 24 hrs treatment. This activity was rapid and mechanism-based since activation of downstream apoptotic signaling 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 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 shows the ability of ABBV-621 to synergize in a panel of pancreatic cancer cell lines with six standard of care agents used in the treatment of pancreatic cancer (gemcitabine, 5-fluorouracil (5FU), erlotinib, paclitaxel, docetaxel, and SN-38 (the active form of irinotecan)) as well as several additional apoptosis inducing anti-cancer agents (navitoclax, venetoclax, and Compound (I)).

BRIEF SUMMARY OF THE INVENTION

[0005] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, in combination with one or more anti-pancreatic cancer agents.

[0006] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with one or more anti-pancreatic cancer agents selected from gemcitabine, 5- fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan.

[0007] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of an anti-pancreatic cancer agent selected from gemcitabine, 5-fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan is administered prior, after, to or concomitantly with a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621.

[0008] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with one or more apoptosis inducing anti-cancer agents selected from navitoclax, venetoclax, and Compound (I).

[0009] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, gemcitabine.

[0010] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, gemcitabine, and with nab-paclitaxel.

[0011] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, docetaxel.

[0012] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, 5-fluorouracil.

[0013] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, erlotinib.

[0014] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, paclitaxel.

[0015] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the anti-pancreatic cancer agent, irinotecan.

[0016] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the apoptosis inducing anti-cancer agent, navitoclax.

[0017] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the apoptosis inducing anti-cancer agent, venetoclax. [0018] The present invention pertains to a method for the treatment of pancreatic cancer comprising the synergistic combination of a therapeutically effective amount of a TRAIL receptor agonist, ABBV-621, with the apoptosis inducing anti-cancer agent, Compound (I).

[0019] The present invention pertains to a method for the treatment of pancreatic 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 the anti- pancreatic cancer agent, gemcitabine.

[0020] The present invention pertains to a method for the treatment of pancreatic 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 the anti- pancreatic cancer agents, gemcitabine and nab-paclitaxel.

[0021] The present invention pertains to a method for the treatment of pancreatic 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- pancreatic cancer agent selected from the group consisting of: paclitaxel and nab-paclitaxel.

[0022] The present invention pertains to a method for the treatment of pancreatic 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 the anti- pancreatic cancer agent, SNA 8.

[0023] The present invention pertains to a method for the treatment of pancreatic 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 the anti- pancreatic cancer agent, docetaxel.

BRIEF DESCRIPTION OF THE DRAWINGS

[0024] FIGURE 1 shows the dose response curves of the combination of ABBV-621 and navitoclax against the BXPC-3 pancreatic cancer cell line. The maximum BLISS score was 46 and the maximum fold ICso was 131. This is representative of a strong synergistic response. [0025] FIGURE 2 shows the dose response curves of the combination of ABBV-621 and navitoclax against the MIA PaCa-2 pancreatic cancer cell line. The maximum BLISS score was 19 and the maximum fold ICso was 4. This is not representative of a synergistic response.

[0026] FIGURE 3 shows the dose response curves of the combination of ABBV-621 and gemcitabine against the SU.86.86 pancreatic cancer cell line. The maximum BLISS score was 21 and the maximum fold ICso was 74. This is representative of a weak synergistic response.

[0027] FIGURE 4 shows the dose response curves of the combination of ABBV-621 and gemcitabine against the DAN-G pancreatic cancer cell line. The maximum BLISS score was 51 and the maximum fold ICso was 351. This is representative of a strong synergistic response.

[0028] FIGURE 5 shows the dose response curves of the combination of ABBV-621 and paclitaxel against the MIA PaCa-2 pancreatic cancer cell line. The maximum BLISS score was 27 and the maximum fold ICso was 25. This is representative of a weak synergistic response.

[0029] FIGURE 6 shows the dose response curves of the combination of ABBV-621 and paclitaxel against the PA-TU-8988T pancreatic cancer cell line. The maximum BLISS score was 58 and the maximum fold ICso was 531. This is representative of a strong synergistic response.

[0030] FIGURE 7 shows the dose response curves of the combination of ABBV-621 and SN- 38 against the BXPC-3 pancreatic cancer cell line. The maximum BLISS score was 29 and the maximum fold ICso was 6290. This is representative of a weak synergistic response.

[0031] FIGURE 8 shows the dose response curves of the combination of ABBV-621 and SN- 38 against the SU.86.86 pancreatic cancer cell line. The maximum BLISS score was 42 and the maximum fold ICso was 257. This is representative of a strong synergistic response.

[0032] FIGURE 9 shows the growth inhibition curves of xenografted human pancreatic carcinoma tumors (patient-derived xenograft model) CTG-0282 by ABBV-621, gemcitabine or docetaxel as single agents or with ABBV-621 in combination with gemcitabine or docetaxel as described in Example 2. As shown, ABBV-621 administered as monotherapy was efficacious. Docetaxel administered as monotherapy or in combination with ABBV-621 compared to ABBV- 621 alone was not efficacious in this model. As shown, gemcitabine as monotherapy or the combination of ABBV-621 with gemcitabine resulted in enhanced tumor growth inhibition. [0033] FIGURE 10 shows the growth inhibition curves of xenografted human pancreatic carcinoma tumors (patient-derived xenograft model) CTG-0289 by ABBV-621, gemcitabine or docetaxel as single agents or with ABBV-621 in combination with gemcitabine or docetaxel as described in Example 2. As shown in Figure 10, the combination of ABBV-621 with docetaxel showed increased efficacy when compared to docetaxel alone. Gemcitabine administered as monotherapy was efficacious. As shown in Figure 10, the combination of ABBV-621 with gemcitabine showed increased efficacy when compared to gemcitabine alone.

[0034] FIGURE 11 shows the growth inhibition curves of xenografted human pancreatic carcinoma tumors (patient-derived xenograft model) CTG-0314 by ABBV-621, gemcitabine or docetaxel as single agents or with ABBV-621 in combination with gemcitabine or docetaxel as described in Example 2. As shown in Figure 11, the combination of ABBV-621 with docetaxel showed synergistic efficacy when compared to docetaxel alone and the lack of efficacy observed for ABBV-621 in this model. Gemcitabine administered as monotherapy was not efficacious in this model. The combination of ABBV-621 with gemcitabine did not show increased efficacy when compared to gemcitabine alone.

[0035] FIGURE 12 shows the growth inhibition curves of xenografted human pancreatic carcinoma tumors (patient-derived xenograft model) CTG-0492 by ABBV-621, gemcitabine or docetaxel as single agents or with ABBV-621 in combination with gemcitabine or docetaxel as described in Example 2. As shown in Figure 12, ABBV-621 administered as monotherapy was not efficacious in this model. Docetaxel administered as monotherapy was efficacious. As shown in Figure 12, the combination of ABBV-621 with docetaxel showed a trend toward increased efficacy when compared to docetaxel alone. Gemcitabine administered as

monotherapy was not efficacious in this model. As shown in Figure 12, the combination of ABBV-621 with gemcitabine showed a synergistic trend toward increased efficacy when compared to gemcitabine alone, which is further highlighted in view of the fact that neither ABBV-621 nor gemicitabine alone were efficacious in this model.

DETAILED DESCRIPTION OF THE INVENTION [0036] 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 ability of ABBV-621 to synergize in a panel of pancreatic cancer cell lines with six standard of care agents used in the treatment of pancreatic cancer (gemcitabine, 5- fluorouracil (5FU), erlotinib, paclitaxel docetaxel, and SN-38 (the active form of irinotecan)) as well as several apoptosis inducing anti-cancer agents (navitoclax, venetoclax, and Compound (I)) illustrates a method for the treatment of pancreatic cancer.

[0037] 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 disulfide 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 A-glycosylated, and further post-translationally modifed by changing the A -term i an glutamine to pyroglutamate, as disclosed in US Patent Publication No. 2015/0337027, incorporated herein by reference in its entirety and for all purposes.

[0038] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 one or more anti-pancreatic cancer agents.

[0039] In one embodiment, the present invention relates to a synergistic method for the treatment of pancreatic 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 one or more anti-pancreatic cancer agents selected from gemcitabine, 5-fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan.

[0040] In one embodiment, the present invention relates to a synergistic method for the treatment of pancreatic cancer in a subject who is in need thereof, comprising administering to the subject one or more anti-pancreatic cancer agents selected from gemcitabine, 5-fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan prior to or concomitantly with an effective amount of the TRAIL receptor agonist, ABBV-621. [0041] In one embodiment, the present invention relates to a synergistic method for the treatment of pancreatic cancer in a subject who is in need thereof, comprising administering to the subject one or more anti-pancreatic cancer agents selected from gemcitabine, 5-fluorouracil, erlotinib, paclitaxel, docetaxel, and irinotecan prior to administering an effective amount of the TRAIL receptor agonist, ABBV-621, such that said anti -pancreatic 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. For this purpose,“sufficient time” may constitute 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.

[0042] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, gemcitabine.

[0043] As is known in the art, gemcitabine has a CAS Registry Number of 95058-81-4; has an empirical formula of C 9 H11F2N3O4; a gram molecular weight is 263.20; and is described in GB Patent Publication No. GB2136425A, published September 19, 1984, incorporated herein by reference in its entirety and for all purposes. Use of the term "4-amino-l-[(2i?,4i?,5i?)-3,3- difluoro-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidin-2( l//)-one " encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of gemcitabine or its salts. Pharmaceutical compositions of 4-amino-l-[(2R,,4R ,5R )-3,3-difluoro-4-hydroxy-5- (hydroxy methyl )oxolan-2-yl]pyri mi din-2( l T/j-one include all pharmaceutically acceptable compositions comprising

ancf one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising

3,3-difluoro-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidi n-2( 1 H )-one is GEMZAR

(hydroxy methyl )oxolan-2-yl]pyri mi din-2( l T/j-one as the active ingredient, also referred to as gemcitabine. Vials of GEMZAR ® contain either 200 mg or 1 g of gemcitabine HC1 (expressed as free base) formulated with mannitol (200 mg or 1 g, respectively) and sodium acetate (12.5 mg or 62.5 mg, respectively) as a sterile lyophilized powder. Hydrochloric acid and/or sodium hydroxide may have been added for pH adjustment. [0044] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, gemcitabine, in further combination with nab-paclitaxel.

[0045] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, 5-fluorouracil.

[0046] As is known in the art, 5-fluorouracil has a CAS Registry Number of 51-28-8; has an empirical formula of C4H3FN2O2; a gram molecular weight of 130.08; and is described in US Patent No. 2,802,005 granted August 6, 1957. Use of the term“5-fluorouracil” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of 5-fluorouracil or its salts. Pharmaceutical compositions of 5-fluorouracil include all pharmaceutically acceptable compositions comprising 5-fluorouracil and one or more diluents, vehicles and/or excipients. 5- Fluorouracil is available in two pharmacy bulk vials, 2.5 g/50 mL vial or 5 g/lOO mL vial in water for injection, pH adjusted to 8.6 to 9.4 with sodium hydroxide.

[0047] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, erlotinib.

[0048] As is known in the art, erlotinib has a CAS Registry Number of 183321-74-6; has an empirical formula of C22H23N3O4; a gram molecular weight of 393.44; and is described in US Patent No. USRE41065E1 granted December 29, 2009, incorporated herein by reference in its entirety and for all purposes. Use of the term“A-(3-ethynylphenyl)-6,7-bis(2- methoxyethoxy)quinazolin-4-amine” encompasses (unless otherwise indicated) solvates

(including hydrates) and polymorphic forms of A-(3-ethynylphenyl)-6,7-bis(2- methoxyethoxy)quinazolin-4-amine or its salts. Pharmaceutical compositions of A-(3- ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine include all pharmaceutically acceptable compositions comprising A-(3 -ethynyl phenyl )-6,7-bi s(2-methoxyethoxy )quinazolin-4- amine and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising /V-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-am ine is TARCEVA ® (OSI Pharmaceuticals, LLC). TARCEVA ® tablets for oral administration are available in three dosage strengths containing erlotinib hydrochloride (27.3 mg, 109.3 mg and 163.9 mg) equivalent to 25 mg, 100 mg and 150 mg erlotinib and the following inactive ingredients: lactose monohydrate, hypromellose, hydroxypropyl cellulose, magnesium stearate, microcrystalline cellulose, sodium starch glycolate, sodium lauryl sulfate and titanium dioxide. The tablets also contain trace amounts of color additives, including FD&C Yellow #6 (25 mg only) for product identification.

[0049] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, paclitaxel.

[0050] As is known in the art, paclitaxel has a CAS Registry Number of 33069-62-4 ; has an empirical formula C47H51NO14; and a gram molecular weight of 853.9. U se of the term u (2aR,4S,4aS,6R,9S, 1 lri) 12S, 12aR, l2bri)-6, l2b-bis(acetyloxy)-9-{ [(2 R,3ri)-3-benzamido-2- hy droxy-3 -phenylpropanoyl] oxy } -4, 11 -dihy droxy-4a, 8, 13,13 -tetramethyl-5 -oxo-

2a, 3 ,4,4a, 5,6,9, 10, 11 , 12, 12a, 12b-dodecahydro- 1/7-7, 11 -methanocyclodeca[3 ,4]benzo[ 1 ,2-

/>]oxet- 12-yl benzoate” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of (2a7?,4ri',4ari , ,6i?,9ri , ,l lri , , l2,S , ,l2a7?, l2bri)-6,l2b-bis(acetyloxy)-9-

{[(2/?,3ri)-3-benzamido-2-hydroxy-3-phenylpropanoyl]oxy}- 4, l l-dihydroxy-4a,8, l3,l3- tetramethyl-5-oxo-2a,3,4,4a,5,6,9, l0,l l, l2,l2a,l2b-dodecahydro-l/7-7, l l- methanocyclodeca[3,4]benzo[l,2-b>]oxet-l2-yl benzoate or its salts. Pharmaceutical compositions of ( 2aR,4S,4aS,6R,9S , 1 1 S, 12S\ 12a// 12bS)-6, 12b-bis(acetyloxy)-9- { [(2//3,S)-3-benzamido-2- hy droxy-3 -phenylpropanoyl] oxy } -4, 11 -dihy droxy-4a, 8, 13,13 -tetramethyl-5 -oxo-

2a, 3 ,4,4a, 5,6,9, 10, 11 , 12, 12a, 12b-dodecahydro- 1/7-7, 11 -methanocyclodeca[3 ,4]benzo[ 1 ,2-

/>]oxet- 12-yl benzoate include all pharmaceutically acceptable compositions comprising

(2aR,4S,4aS,6R,9S, 1 1 S, 12S\ 12a// 12bk)-6, 12b-bis(acetyloxy)-9- { [(2//3,S)-3-benzamido-2- hy droxy-3 -phenylpropanoyl] oxy } -4, 11 -dihy droxy-4a, 8, 13,13 -tetramethyl-5 -oxo-

2a, 3 ,4,4a, 5,6,9, 10, 11 , 12, 12a, 12b-dodecahydro- 1/7-7, 11 -methanocyclodeca[3 ,4]benzo[ 1 ,2-

/>]oxet- 12-yl benzoate and one or more diluents, vehicles and/or excipients. Paclitaxel Injection is a clear, colorless to slightly yellow viscous solution. It is supplied as a nonaqueous solution intended for dilution with a suitable parenteral fluid prior to intravenous infusion. Paclitaxel Injection is available in 30 mg (5 mL), 100 mg (16.7 mL), and 300 mg (50 mL) multidose vials. Each mL of sterile nonpyrogenic solution contains 6 mg paclitaxel, 527 mg of purified

Cremophor ® EL* (polyoxyethylated castor oil) and 49.7% (v/v) dehydrated alcohol, LTSP.

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 noncrystalline, amorphous state. ABRAXANE ® is supplied as a white to yellow, sterile, lyophilized powder for reconstitution with 20 mL of 0.9% Sodium Chloride Injection, LTSP 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.

[0051] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, docetaxel.

[0052] 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-erocn- 1 ,2a,4,7b, 10b, 13a-hexahydroxytax- 1 1 -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.

[0053] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 anti-pancreatic cancer agent, irinotecan.

[0054] As is known in the art, irinotecan has a CAS Registry Number 97682-44-5 ; has an empirical formula C 33 H 38 N4O 6 ; and a gram molecular weight of 586.68; and is described in Japanese Patent Publication JP60019790 published January 31, 1985, incorporated herein by reference in its entirety and for all purposes. Lise of the term“(4ri)-4, l l-diethyl-4-hydroxy- 3, l4-dioxo-3,4, l2,l4-tetrahydro-li7-pyrano[3',4':6,7]indolizino[l,2-/>]q uinolin-9-yl [1,4 - bipiperidine]-l'-carboxylate” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of (4ri)-4,l l-diethyl-4-hydroxy-3,l4-dioxo-3,4,l2, l4- tetrahydro-li7-pyrano[3',4':6,7]indolizino[l,2-/>]quinoli n-9-yl [l,4'-bipiperidine]-l'-carboxylate or its salts. Pharmaceutical compositions of (4ri)-4,l l-diethyl-4-hydroxy-3, l4-dioxo-3,4,l2, l4- tetrahydro-li7-pyrano[3',4':6,7]indolizino[l,2-/>]quinoli n-9-yl [l,4'-bipiperidine]-l'-carboxylate include all pharmaceutically acceptable compositions comprising (4S')-4, 1 1 -diethyl-4-hydroxy- 3, l4-dioxo-3,4, l2,l4-tetrahydro-li7-pyrano[3',4':6,7]indolizino[l,2-/bvq]in olin-9-yl [1,4 - bipiperidine]-l'-carboxylate and one or more diluents, vehicles and/or excipients. One example of a pharmaceutical composition comprising“(4ri)-4,l l-diethyl-4-hydroxy-3, l4-dioxo-3,4, l2,l4- tetrahydro-liT-pyrano[3',4':6,7]indolizino[l,2-B quinolin-9-yl [l,4'-bipiperidine]-l'-carboxylate is CAMPTOSAR ® (Pfizer Inc ). 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 is 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“(4ri)- 4,1 l-diethyl-4-hydroxy-3,l4-dioxo-3,4,l2,l4-tetrahydro-liT-pyra no[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-hydroxyethyl) piperazin-l-yl]ethanesulfonic acid (HEPES) as a buffer 4.05 mg/mL and sodium chloride as an isotonicity reagent 8.42 mg/mL.

[0055] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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.

[0056] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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 selected from navitoclax, venetoclax, and Compound (I).

[0057] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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, navitoclax. [0058] As is known in the art, navitoclax has a CAS Registry Number of 923564-51-6 ; has an empirical formula C47H55CIF3N5O6S3; and a gram molecular weight of 974.61; and is described in US Patent No. 7390799 granted June 24, 2008, incorporated herein by reference in its entirety and for all purposes. Use of the term“4-{4-[(4'-chloro-4,4-dimethyl-3,4,5,6-tetrahydro[l, l'- biphenyl]-2-yl)methyl]piperazin-l-yl}-/V-[4-{[(2i?)-4-(morph olin-4-yl)-l-(phenylsulfanyl)butan- 2-yl]amino}-3-(trifluoromethanesulfonyl)benzene-l-sulfonyl]b enzamide” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of 4-{4-[(4'-chloro-4,4- dimethyl-3 ,4,5,6-tetrahydro[ 1 , 1 '-biphenyl]-2-yl)methyl]piperazin- 1 -yl } -N-[4- { [(2i?)-4- (morpholin-4-yl)-l-(phenylsulfanyl)butan-2-yl]amino}-3-(trif luoromethanesulfonyl)benzene-l- sulfonyljbenzamide or its salts. Pharmaceutical compositions of 4-{4-[(4'-chloro-4,4-dimethyl- 3,4,5,6-tetrahydro[l,l'-biphenyl]-2-yl)methyl]piperazin-l-yl }-N-[4-{[(2i?)-4-(morpholin-4-yl)-l- (phenylsulfanyl)butan-2-yl]amino}-3-(trifluoromethanesulfony l)benzene-l-sulfonyl]benzamide include all pharmaceutically acceptable compositions comprising 4-{4-[(4'-chloro-4,4-dimethyl- 3,4,5,6-tetrahydro[l,l'-biphenyl]-2-yl)methyl]piperazin-l-yl }-N-[4-{[(2i?)-4-(morpholin-4-yl)-l- (phenylsulfanyl)butan-2-yl]amino}-3-(trifluoromethanesulfony l)benzene-l-sulfonyl]benzamide and one or more diluents, vehicles and/or excipients.

[0059] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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, venetoclax.

[0060] As is known in the art, venetoclax has a CAS Registry Number of 1257044-40-8 ; has an empirical formula C45H50CIN7O7S; and a gram molecular weight of 868.44; and is described in PCT Patent Application Publication WO 2010/138588 published December 2, 2010,

incorporated herein by reference in its entirety and for all purposes. Use of the term“4-{4-[(4 - chloro-5,5-dimethyl-3,4,5,6-tetrahydro[l,l'-biphenyl]-2-yl)m ethyl]piperazin-l-yl}-A-(3-nitro-4- {[(oxan-4-yl)methyl]amino}benzene-l-sulfonyl)-2-[(U/-pyrrolo [2,3-Z>]pyridin-5- yl)oxy]benzamide” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of 4-{4-[(4'-chloro-5,5-dimethyl-3,4,5,6-tetrahydro[l, l'-biphenyl]-2- yl)methyl]piperazin-l-yl}-A-(3-nitro-4-{[(oxan-4-yl)methyl]a mino}benzene-l-sulfonyl)-2-[(U7- pyrrolo[2,3-Z ]pyridin-5-yl)oxy]benzamide or its salts. Pharmaceutical compositions of 4-{4-[(4'- chloro-5,5-dimethyl-3,4,5,6-tetrahydro[l,l'-biphenyl]-2-yl)m ethyl]piperazin-l-yl}-A-(3-nitro-4- {[(oxan-4-yl)methyl]amino}benzene-l-sulfonyl)-2-[(li7-pyrrol o[2,3-/>]pyridin-5- yl)oxy]benzamide include all pharmaceutically acceptable compositions comprising 4-{4-[(4'- chloro-5,5-dimethyl-3,4,5,6-tetrahydro[l,r-biphenyl]-2-yl)me thyl]piperazin-l-yl}-/V-(3-nitro-4- {[(oxan-4-yl)methyl]amino}benzene-l-sulfonyl)-2-[(li7-pyrrol o[2,3-/>]pyridin-5- yl)oxy]benzamide and one or more diluents, vehicles and/or excipients. Venetoclax tablets for oral administration are supplied as pale yellow or beige tablets that contain 10, 50, or 100 mg venetoclax as the active ingredient. Each tablet also contains the following inactive ingredients: copovidone, colloidal silicon dioxide, polysorbate 80, sodium stearyl fumarate, and calcium phosphate dibasic. In addition, the 10 mg and 100 mg coated tablets include the following: iron oxide yellow, polyvinyl alcohol, polyethylene glycol, talc, and titanium dioxide. The 50 mg coated tablets also include the following: iron oxide yellow, iron oxide red, iron oxide black, polyvinyl alcohol, talc, polyethylene glycol and titanium dioxide.

[0061] In one embodiment, the present invention relates to a method for the treatment of pancreatic 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).

[0062] As is known in the art, Compound (I) has a CAS Registry Number of 1430845-59-2; has an empirical formula C43H49N7O4S; and a gram molecular weight of 759.96; and is described in PCT Patent Application Publication WO 2013/055897 published April 18, 2013, incorporated herein by reference in its entirety and for all purposes. Use of the term“6-{8-[(l,3-benzothiazol- 2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(li7)-yl}-3-[l-({3,5 -dimethyl-7-[2- (methylamino)ethoxy]adamantan- 1 -yl }methyl)-5-methyl- liT-pyrazol-4-yl]pyridine-2-carboxylic acid” encompasses (unless otherwise indicated) solvates (including hydrates) and polymorphic forms of 6-{8-[(l,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinol in-2(li7)-yl}-3-[l-({3,5- dimethyl-7-[2-(methylamino)ethoxy]adamantan- 1 -yl }methyl)-5-methyl- liT-pyrazol-4- yl]pyridine-2-carboxylic acid or its salts. Pharmaceutical compositions of 6-{8-[(l,3- benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(l//)-y l}-3-[l-({3,5-dimethyl-7-[2- (methylamino)ethoxy]adamantan- 1 -yl }methyl)-5-methyl- liT-pyrazol-4-yl]pyridine-2-carboxylic acid include all pharmaceutically acceptable compositions comprising 6-{8-[(l,3-benzothiazol-2- yl)carbamoyl]-3,4-dihydroisoquinolin-2(l//)-yl}-3-[l-({3,5-d imethyl-7-[2- (methylamino)ethoxy]adamantan- 1 -yl }methyl)-5-methyl- l//-pyrazol-4-yl]pyridine-2-carboxylic acid and one or more diluents, vehicles and/or excipients.

[0063] 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.

[0064] 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.

[0065] 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.

[0066] So that the disclosure may be more readily understood, select terms are defined below.

DEFINITIONS

[0067] The terms "treat", "treating" and "treatment" refer to a method of alleviating or abrogating a disease and/or its attendant symptoms.

[0068] 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. [0069] The terms“patient” and“subject” are used herein interchangeably.

[0070] 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.

[0071] "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 with anti-cancer agents to treat or prevent pancreatic cancer at a reasonable benefit/risk ratio applicable to any medical treatment.

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

[0072] “Specificity” refers to the ability of a binding protein to selectively bind an antigen or receptor.

[0073] 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.

[0074] 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.

[0075] 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.

[0076] 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.

[0077] “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).

[0078] 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.

[0079] 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). [0080] “Navitoclax” is 4-{4-[(4'-chloro-4,4-dimethyl-3,4,5,6-tetrahydro[l, r-biphenyl]-2- yl)methyl]piperazin-l-yl}-/V-[4-{[(2i?)-4-(morpholin-4-yl)-l -(phenylsulfanyl)butan-2-yl]amino}-

3-(trifluoromethanesulfonyl)benzene-l-sulfonyl]benzamide.

[0081] “Venetoclax” is 4-{4-[(4'-chloro-5,5-dimethyl-3,4,5,6-tetrahydro[l, r-biphenyl]-2- yl (methyl ]piperazin- l -yl }-A-(3-nitro-4- i [(oxan-4-yl (methyl ]ami no } benzene- 1 -sulfonyl(-2-[( 1 /7- pyrrolo[2,3-/ ]pyridin-5-yl)oxy]benzamide

[0082] “Irinotecan” is (4S'(-4, 1 1 -di ethyl -4-hy droxy-3 , 14-di oxo-3 ,4, 12, 14-tetrahy dro- 177- pyrano[3',4':6,7]indolizino[l,2-b ]quinolin-9-yl [l,4'-bipiperidine]-l'-carboxylate.

[0083] “SN-38” is (4S)-4, l 1 -diethyl-4, 9-dihydroxy-l/7-pyrano[3',4':6,7]indolizino[l, 2-

7>] quinoline-3, 14(4/7, l2/7)-dione. SN-38 is the active metabolite of irinotecan and is formed by hydrolysis of irinotecan by carboxylesterases, mainly in the liver. In vitro assays necessitate the use of SN-38 to explore the efficacy of irinotecan.

[0084] “Gemcitabine” is 4-amino-l-[(2i?,4i?,5i?)-3,3-difluoro-4-hydroxy-5- (hydroxy methyl )oxolan-2-yl]pyri mi din-2( l //)-one.

[0085] “Erlotinib” is A-(3-ethynyl phenyl )-6,7-bis(2-methoxyethoxy)quinazolin-4-amine.

[0086] “Paclitaxel” is (2aiR,4s , ,4a,S , ,6R,,9,S , , l l S , ,l2S , , l2aR, l2b,S)-6, l2b-bis(acetyloxy)-9- {[(2i?,3S)-3-benzamido-2-hydroxy-3-phenylpropanoyl]oxy}-4, l l-dihydroxy-4a,8, l3,l3- tetramethyl-5-oxo-2a,3,4,4a,5,6,9, l0,l l, l2,l2a,l2b-dodecahydro-l/7-7, 11- methanocyclodeca[3,4]benzo[l,2-/ ]oxet-l2-yl benzoate.

[0087] “Docetaxel” is (2A,3S')-N -carboxy-3 -phenyl isoserine, 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.

[0088] “5-Fluorouracil” is 5-fluoropyrimidine-2, 4(1HH, 3H )-dione.

[0089] “Compound (I)” is 6-{8-[(l,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinol in- 2(l/7)-yl}-3-[l-({3,5-dimethyl-7-[2-(methylamino)ethoxy]adam antan-l-yl}methyl)-5-methyl- l //-pyrazol-4-yl]pyridine-2-carboxylic acid, an inhibitor of Bcl-xL.

Compound (I)

[0090] 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.

[0091] 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.

[0092] 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.

BRIEF DESCRIPTION OF THE SEQUENCE LISTING

[0093] Incorporated herein by reference in its entirety is a Sequence Listing entitled, “ABV12441WO01 ST25”, comprising SEQ ID NO: 1, 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 7 KB in size.

EXAMPLES

Example 1: Treatment of Pancreatic Cancer Cell Lines with ABBV-621 and in

Combination with Standard of Care Agents and with Apoptosis Inducing Anti-Cancer Agents

Materials and Methods

[0094] Cell Culture : All cell lines were sourced from either American Type Culture Collection (ATCC) or Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) and obtained through the Abb Vie cancer core cell line facility which verifies the identity of each cell line with short tandem repeat (STR) profiling (Table 2). All reagents used for cell culture are listed in Table 3. Frozen stocks of cells were thawed at 37 °C and transferred to 15 mL conical tubes containing 10 mL of indicated media (cell line dependent). Cells were collected by centrifugation for 3 minutes at 500 x g. The media was removed; the cells were re-suspended in 5 mL of media, and transferred to Corning T75 flasks containing 10 mL media. Cells were incubated at 37 °C/5% CO2 in a humidified incubator until ~ 75% confluent at which time the media was removed and the cells were detached from the plate in 3 mL of Trypsin-EDTA, and transferred to two T175 flasks containing the 30 mL of the appropriate media. Cells were incubated at 37 °C/5% CO2 in a humidified incubator until ~ 75% confluent, detached from the plate using 5 mL Trypsin-EDTA, and transferred to 15 mL conical tubes with 7 mL of media. Cells were then counted on a Beckman Coulter Vi-CELL™, and 3.6 x 10 6 viable cells were diluted to 59 mL media in a 100 mL reservoir. Cells were plated at 25 pL/well in 384 well tissue culture plates (1500 cells/well). Plates were centrifuged at 100 x g for 1 minute in a swinging bucket rotor to ensure the cells were on the bottom, and then incubated 18-24 hours at 37 °C/5% CO2 in a humidified incubator.

Abbreviations:

[0095] DMEM for Dulbecco’s modified Eagle medium; FBS for fetal bovine serum; IMDM for Iscove’s modification of DMEM; IMEM for improved minimum essential medium; MEM for minimum essential medium; Pen Strep for penicillin streptomycin; RPMI for Roswell Park Memorial Institute medium; and Trypsin-EDTA for trypsin-ethylenediaminetetraacetic acid.

Table 2: Pancreatic cancer cell lines used in the in vitro cell cytotoxicity assays

Table 3: Reagents used for Cell culture

[0096] Dose Response : Compounds used, and the maximum concentration for each, are described in Table 4. Half-log serial dilutions of ABBV-621 (10 dilutions and media control) and compounds (9 dilutions and media control) starting at seven times the indicated maximum concentration in Table 4 were made in the cell line specific media. Test compounds were added at 5 pL/well. After compound addition, 5 pL/well of ABBV-621 was then added. The final volume in each well was 35 pL with 5 pL of each compound. Plates were centrifuged for 1 minute at 100 x g to ensure compounds were in the media and then incubated at 37 °C/5% CO2.

Table 4: Compounds used along with top dose for each for treatment of pancreatic cancer cell lines

[0097] Viability determination : Following incubation for approximately 24 hours, 25 pL of CellTiter Glo ® reagent (Promega, Madison WI, Cat # G7571) was added to all wells. The plates were incubated at room temperature for 15-45 minutes and assayed for luminescence on a Biotek ® Synergy™ Neo2 plate reader.

[0098] Calculations. Synergy was determined based on the Bliss Independence Model (Borisy et al. Proc Natl Acad Sci USA 2003; 100: 7977-7982). ICso values were determined using GraphPad Prism ® using the curve fitting algorithm based on log(inhibitor) versus normalized response using a variable slope. Samples were normalized to the average of the two media controls on each plate. Fold ICso was determined by taking the calculated ICso values at each combination, and comparing these values to the single agent ABBV-621 ICso. If the single agent ABBV-621 treatment ICso was > 20 nM, the ICso of the ABBV-621 treatment was set to 20 nM in order to avoid extrapolated data. If all combinations produced an ABBV-621 ICso value > 20 nM, then the fold ICso was set to zero.

Results:

Single agent activity: [0099] Single agent activity of ABBV-621, as defined by an IC50 of <20 nM, was observed in 9 of the 14 cell lines. Of the 9 sensitive cell lines, 6 cell lines showed sub-nanomolar sensitivity. The IC50 for ABBV-621 along with the % growth inhibition of ABBV-621 alone or in combination with each test compound is shown in Table 5.

[00100] The single agent activity of each test compound at 24 hour treatment was also determined (Table 6). A compound was considered to have single agent activity if the IC50 was less than the maximum concentration used. Compound (I) and SN-38 showed single agent activity in 5 or 6 of the 14 pancreatic cell lines, respectively. Navitoclax showed single agent activity in 3 of the 14 cell lines. 5-Fluorouracil (5FU) and erlotinib showed weak activity in cell lines DAN-G and BXPC3, respectively. No single agent activity was observed for gemcitabine, venetoclax, or paclitaxel in any of the cell lines.

Table 5: IC50 of ABBV-621 and percent growth inhibition of ABBV-621 alone or in combination with each test compound after 24 hour incubation.

Table 6: Single agent activity (mM) for all test compounds after 24 hour incubation.

Combination activity:

[00101] The percent growth inhibition values resulting from ABBV-621 treatment in

combination with each test compound are listed in Table 5. Whereas the change in growth inhibition is a useful parameter to understand combination activity, it is difficult to use this alone to assess synergy since many of the cell lines had very high growth inhibition (>90%) with ABBV-621 alone. In this case, the change in growth inhibition could appear very small, but the amount of ABBV-621 necessary to attain the growth inhibition could be very different when used in combination. Therefore, synergy was defined based on a combination of the maximum BLISS score and fold change in ICso for each combination/cell line. Cutoffs were assigned for both BLISS score and fold change in ICso (Table 7). Points were assigned based on the cutoffs and added to come up with a final objective determination of strong synergy (Table 8), defined as strong synergy, weak synergy and no synergy. Using these criteria, out of 112 total combination/cell lines tested, 59 of the tests showed either weak synergy or strong synergy whereas 53 showed no significant synergy. Table 9 shows the final assessment of synergy for each cell line and combination.

[00102] Of the standard of care agents, the combination of ABBV-621 with SN-38 showed strong synergy in 11 of the 14 cell lines and weak synergy in 2 of the remaining cell lines. Only one cell line (HP AC) showed no synergy with SN-38 using the objective criteria. This particular cell line was sensitive to ABBV-621 as a single agent. Representative dose response curves are illustrated in FIGURE 7 and FIGURE 8 for the combination of ABBV-621 with SN-38 in the cell lines BXPC-3 and SU.86.86, respectively, wherein weak and strong synergies are shown, respectively.

[00103] Strong synergy was observed with the gemcitabine/ABBV-62l combination in DAN-G and CFPAC-l while weak synergy was observed in AsPC-l and Su.86.86. Of these cell lines, two (AsPC-l, DAN-G) are relatively resistant to ABBV-621 as a single agent whereas the other two are sensitive (Su.86.86, CFPAC-l). Furthermore, whereas none of the cell lines were sensitive to gemcitabine based on the criteria of ICso < maximum dose (1 mM) in 24 hours, it was clear that there was some single agent activity in three of the four cell lines (SU.86.86, DAN-G and CFPAC-l). Representative dose response curves are illustrated in FIGURE 3 and FIGURE 4 for the combination of ABBV-621 with gemcitabine in the cell lines SU.86.86 and DAN-G, respectively, wherein weak and strong synergies are shown, respectively.

[00104] Paclitaxel showed strong synergy with ABBV-621 in two cell lines (PA-TU-8688T and HPAF-II) and weak synergy in three cell lines (DAN-G, MIA PaCa-2 and Panc-l).

Interestingly, all of these cell lines except PA-TU-8688T are relatively resistant to ABBV-621 as a single agent (ICso >1 nM). Again, whereas none of these cell lines are regarded as sensitive to Paclitaxel (ICso <1 mM in 24 hour treatment), there were signs of single agent activity in several of the cell lines. Representative dose response curves are illustrated in FIGURE 5 and FIGURE 6 for the combination of ABBV-621 with paclitaxel in the cell lines Mia PaCa-2 and PA-Tu- 8988T, respectively, wherein weak and strong synergies are shown, respectively.

[00105] No cell line showed strong synergy with the combination of ABBV-621 and either 5- fluorouracil or erlotinib, although a few cell lines showed weak synergy with either combination. For 5-fluorouracil, two cell lines show signs of single agent activity (BXPC-3 and SU.86.86) whereas the third cell line (DAN-G) showed very little single agent activity. In contrast, none of the four cell lines (BXPC-3, AsPC-l, Capan-2 and HPAF-II) exhibiting synergy with the ABBV- 621 and erlotinib showed signs of single agent activity with erlotinib.

[00106] For the anti-apoptotic agents, the combination of navitoclax with ABBV-621 and the Bcl-xL inhibitor, Compound (I), with ABBV-621 showed synergy in 12 of the 14 cell lines. Not surprisingly, the two agents showed synergy in 11 of the same cell lines. PA-TU-8988T and PA- TU-8988S showed synergy with navitoclax and no synergy with the Bcl-xL inhibitor, Compound (I), using the objective criteria. In contrast, the Bcl-xL inhibitor, Compound (I), showed weak synergy in HP AC whereas navitoclax did not. This cell line demonstrated single agent activity with the Bcl-xL inhibitor, Compound (I), and the observed synergy appears to be driven by the single agent activity. Representative dose response curves are illustrated in FIGURE 1 and FIGURE 2 for the combination of ABBV-621 with navitoclax in the cell lines BXPC-3 and MIA PaCa-2, respectively, wherein strong and no synergies are shown, respectively.

[00107] In the specific cell lines tested, none of the them had showed strong synergy, as defined herein, with the combination of ABBV-621 with either venetoclax, however, several did show weak synergy. In the 5 cell lines showing weak synergy with venetoclax, only the highest concentration (10 mM) showed a different dose response than the ABBV-621 only control. Higher concentrations of venetoclax did appear to be generally cytotoxic in a number of cell lines so it is difficult to determine whether the observed synergy is simply a function of cytotoxicity from venetoclax.

Table 7: Cutoff values and points for BLISS and fold IC50 calculations

Table 8: Objective criteria for determination of synergy

Conclusions

[00108] Cell cytotoxicity assays were performed on 14 pancreatic cancer cell lines using ABBV-621 in combination with 6 standard of care agents for pancreatic cancer and 3 apoptosis inducing anti-cancer agents. Surprisingly, substantial synergy was observed in several cell lines with several of the tested combinations. Significantly, SN-38, the active form of irinotecan, showed synergistic activity with ABBV-621 in 13 of the 14 cell lines tested. Paclitaxel and gemcitabine also both showed strong synergy with ABBV-621 in several of the cell lines. It is interesting to note that both of these agents had at least two cell lines with substantial synergistic activity. The remaining standard of care agents, 5-fluorouracil and erlotinib, did not demonstrate substantial synergy in any of the cell lines but did show weak synergy in several cell lines.

[00109] Since ABBV-621 induces apoptosis through both the extrinsic and intrinsic apoptosis pathways, the inventors sought to evaluate the combination of ABBV-621 with agents that interfere with the intrinsic anti-apoptotic proteins. The apoptosis inducing agents tested included navitoclax, an inhibitor of both Bcl-xL and Bcl-2; venetoclax, a specific inhibitor of Bcl-2; and Compound (I), a specific inhibitor of Bcl-xL. Of these, the agents that inhibited Bcl-xL alone (Compound (I)) or in combination with Bcl-2 inhibition (navitoclax) showed strong synergy in 9 of the 14 cell lines. This would suggest that the many of the pancreatic cell lines are partially dependent on Bcl-xL when the intrinsic pathway is activated by ABBV-621.

[00110] The combination activity was tested after a 24 hour incubation using a co-treatment.

For most of the test compounds, very little single agent activity was observed under these conditions. Since the activity of several of these compounds (5-fluorouracil, gemcitabine, paclitaxel and SN-38) is cell cycle dependent, additional single agent activity would likely be observed with longer incubations. Therefore, it is likely that additional synergistic or additive effects may be observed if the cells were pre-incubated with the test compound prior to addition of ABBV-621 or if the co-treatment was allowed to continue for longer time periods. The fact that synergy is observed with these compounds at 24 hours would suggest cell cycle-independent changes are occurring that either promote apoptosis or block inhibition of apoptosis (i.e., agents that sensitize the cells for apoptosis) upon treatment with ABBV-621. Example 2: Growth Inhibition of Xenografted Human Patient Derived Pancreatic

Carcinoma Tumors by ABBV-621 in Combination with Docetaxel or Gemcitabine

Materials and Methods

Mice and Husbandry

[00111] Pancreatic patient-derived xenograft (PDX) studies were performed. 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

[00112] 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 (mm 3 ) 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

[00113] 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. Docetaxel and gemcitabine were obtained and formulated in phosphate buffered saline pH 7.2.

Statistics

[00114] 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

[00115] Fresh tumor tissue was cut into fragments and implanted. All tumors were size matched at approximately 200-250 mm 3 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 W 2 /2. Mice were euthanized when tumor volume reached up to 2,000 mm 3 or skin ulcerations occurred.

Experimental Conditions

[00116] CTG-0282, CTG-0289, CTG-0314 or CTG-0492 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 Excel (Microsoft Office). Details of the treatments and route of administration are described in Tables 10.

Results and Discussion

[00117] Efficacy of ABBV-621 was determined in combination with docetaxel and gemcitabine in the CTG-0282 pancreatic patient-derived xenograft model. Agents were administered as shown in Table 10. ABBV-621 administered as monotherapy at 3 mg/kg Q7Dx3 was efficacious with TGI of 64% (p<0.05). Docetaxel administered as monotherapy at 50 mg/kg QDxl or in combination with ABBV-621 compared to ABBV-621 alone was not efficacious in this model as shown in Figure 9. With the exception of the CTG-0282 mice that were treated with ABBV-621 in combination with gemcitabine, all treatments groups were well -tolerated and no significant body weight loss was observed. For the mice administered with the ABBV-621 and gemcitabine combination in the CTG-0282 model, 4 out of 5 mice had died at an early time point within the study. Accordingly, only data for the one surviving mouse is plotted in Figure 9 for that specific treatment group.

[00118] Efficacy of ABBV-621 was determined in combination with docetaxel and gemcitabine in the CTG-0289 pancreatic patient-derived xenograft model. Agents were administered as shown in Table 10. ABBV-621 administered as monotherapy at 3 mg/kg Q7Dx3 was efficacious with TGI of 41% (NS). Docetaxel administered as monotherapy at 50 mg/kg QDxl was efficacious with TGI of 91% (p<0.005). As shown in Figure 10, the combination of ABBV-621 with docetaxel showed increased efficacy when compared to docetaxel alone by TGI (66%; p<0.0l). Gemcitabine administered as monotherapy at 80 mg/kg Q3Dx4 was efficacious with TGI of 59% (NS). As shown in Figure 10, the combination of ABBV-621 with gemcitabine showed increased efficacy when compared to gemcitabine alone by TGD (46%; p<0.05). All treatments groups were well tolerated and no significant body weight loss was observed.

[00119] Efficacy of ABBV-621 was determined in combination with docetaxel and gemcitabine in the CTG-0314 pancreatic patient-derived xenograft model. Agents were administered as shown in Table 10. ABBV-621 administered as monotherapy at 3 mg/kg Q7Dx3 was not efficacious in this model. Docetaxel administered as monotherapy at 50 mg/kg QDxl was efficacious with TGI of 33% (p<0.05). As shown in Figure 11, the combination of ABBV-621 with docetaxel showed synergistic efficacy when compared to docetaxel alone by TGI (74%; p<0.05) and the lack of efficacy observed for ABBV-621 in this model. Gemcitabine

administered as monotherapy at 80 mg/kg Q3Dx4 was not efficacious in this model. The combination of ABBV-621 with gemcitabine did not show increased efficacy when compared to gemcitabine alone. All treatments groups were well tolerated and no significant body weight loss was observed.

[00120] Efficacy of ABBV-621 was determined in combination with docetaxel and gemcitabine in the CTG-0492 pancreatic patient-derived xenograft model. Agents were administered as shown in Table 10. ABBV-621 administered as monotherapy at 3 mg/kg Q7Dx3 was not efficacious in this model. Docetaxel administered as monotherapy at 50 mg/kg QDxl was efficacious with TGI of 42% (NS). As shown in Figure 12, the combination of ABBV-621 with docetaxel showed a trend toward increased efficacy when compared to docetaxel alone by TGI (39%; NS) and TGD (16%; NS). Gemcitabine administered as monotherapy at 80 mg/kg Q3Dx4 was not efficacious in this model. As shown in Figure 12, the combination of ABBV-621 with gemcitabine showed a synergistic trend toward with increased efficacy when compared to gemcitabine alone by TGI (53%; NS) and TGD (29%; NS), which is further highlighted by in view of the fact that neither ABBV-621 nor gemicitabine alone were efficacious in this model. All treatments groups were well tolerated and no significant body weight loss was observed.

Table 10: Study Design