Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR COMBINATION IMMUNOTHERAPY
Document Type and Number:
WIPO Patent Application WO/2016/172249
Kind Code:
A1
Abstract:
Methods and compositions for generating immune responses using adenovirus vectors that allow multiple vaccinations or in combination with other therapy and vaccinations in individuals with preexisting immunity to adenovirus are provided.

Inventors:
JONES FRANK R (US)
GABITZSCH ELIZABETH (US)
LATCHMAN YVETTE (US)
RICE ADRIAN (US)
Application Number:
PCT/US2016/028496
Publication Date:
October 27, 2016
Filing Date:
April 20, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ETUBICS CORP (US)
International Classes:
C07K14/82; G01N33/574
Domestic Patent References:
WO2014043518A12014-03-20
WO2014031178A12014-02-27
WO1996002555A11996-02-01
WO1999033488A21999-07-08
Foreign References:
US20100055069A12010-03-04
US20130251741A12013-09-26
US20100209386A12010-08-19
US20050037439A12005-02-17
US20040091995A12004-05-13
US6063622A2000-05-16
US6451596B12002-09-17
US6057158A2000-05-02
US6083750A2000-07-04
US8298549B22012-10-30
USPP60158585P
US7009042B12006-03-07
US5466468A1995-11-14
US4436727A1984-03-13
US4877611A1989-10-31
US4866034A1989-09-12
US4912094A1990-03-27
US0600820A1898-03-15
US5856462A1999-01-05
US5725871A1998-03-10
US5780045A1998-07-14
Other References:
PHILIPS G. K ET AL., INT. IMMUNOL., vol. 27, 2014, pages 39 - 46
SMITHWATERMAN, ADD. APL. MATH, vol. 2, 1981, pages 482
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PEARSONLIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
LAUER ET AL., J. GEN. VIROL., vol. 85, 2004, pages 2615 - 25
LEZA ET AL., J. VIROL., 1988, pages 3003 - 13
MIRALLES ET AL., J. BIO CHEM., vol. 264, no. 18, 1983, pages 10763 - 72
SKEIKY ET AL., INFECTION AND IMMUN., vol. 67, 1999, pages 3998 - 4007
"GenBank", Database accession no. CR542051.1
"GeneBank", Database accession no. AY358337.1
JNCI, vol. 96, 2004, pages 1035 - 1038,1570-1580
J CLIN ONCOL, vol. 17, 1999, pages 3461 - 3467
J CLIN ONCOL, vol. 26, 2008, pages 1148 - 1159
JNCI, vol. 92, 2000, pages 1534 - 1535
See also references of EP 3286213A4
Attorney, Agent or Firm:
WANG, Qiong (650 Page Mill RoadPalo Alto, California, US)
Download PDF:
Claims:
CLAIMS

1. A method of enhancing an immune response in an individual in need thereof, the method comprising:

administering to the individual a first pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen; and

administering to the individual an immune checkpoint inhibitor.

2. The method of claim 1, wherein the HPV antigen is HPV-E6, HPVE7, or a combination thereof.

3. The method of claim 1 or 2, wherein the HPV antigen is a non-oncogenic HPV antigen.

4. The method of any of claims 1-3, wherein the HPV antigen is a modified HPV E6 antigen having an amino acid sequence set forth in SEQ ID NO: 17, a modified HPV E7 antigen having an amino acid sequence set forth in SEQ ID NO: 18, or a combination thereof.

5. The method of any of claims 1-4, wherein the HPV antigen is a modified HPV E6 antigen that has one or more mutations at positions 26, 98, or 106 of SEQ ID NO: 17, a modified HPV E7 antigen that a mutation at position 86 of SEQ ID NO: 18, or a combination thereof.

6. The method of any of claims 1-5, wherein the nucleic acid sequence has a region at least 80% identical to positions 123-545 of SEQ ID NO: 19, positions 602-895 of SEQ ID NO:9, or a combination thereof.

7. The method of any of claims 1-6, wherein the nucleic acid sequence has at least 80% identity to SEQ ID NO: 19.

8. The method of any of claims 1-7, wherein the nucleic acid sequence has at least 80% identity to SEQ ID NO:20.

9. The method of any of claims 1-8, wherein the immune checkpoint inhibitor targets PD1, PDLl, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, or CD244.

10. The method of any of claims 1-9, wherein the immune checkpoint inhibitor targets PD1 or PDLl .

11. The method of any of claims 1-10, wherein the immune checkpoint inhibitor is an anti-PD-1 or anti-PD-Ll antibody.

12. The method of any of claims 1-11, wherein the immune checkpoint inhibitor is an anti-PD- Ll antibody.

13. The method of any of claims 1-12, wherein the immune checkpoint inhibitor is avelumab.

14. The method of any of claims 1-13, wherein the replication-defective adenovirus vector has a deletion in an early 2b (E2b) gene region.

15. The method of any of claims 1-14, wherein the replication-defective adenoviral vector further comprises a deletion in an early 1 (El) gene region, a deletion in an early 3 (E3) gene region, a deletion in an early 4 (E4) gene region, or a combination thereof.

16. The method of any of claims 1-15, wherein the method is further defined as a method of treating an HPV infection or an HPV-associated disease in an individual in need thereof.

17. The method of any of claims 1-16, wherein the individual has an HPV infection or an HPV- associated disease.

18. The method of any of claims 1-17, wherein the HPV-associated disease is HPV associated head and neck squamous cell carcinoma (HNSCC), oropharyngeal and tonsillar cancer, anal cancer, or cervical cancer.

19. The method of any of claims 1-18, wherein the individual is a human.

20. The method of any of claims 1-19, wherein the individual has pre-existing immunity to Ad5.

21. The method of any of claims 1-20, wherein the administration is repeated at every three weeks.

22. The method of any of claims 1-21, wherein the pharmaceutical composition comprises at least 1011 adenovirus vectors.

23. A method of enhancing an immune response in an individual in need thereof, the method comprising administering to the individual a first replication-defective vector comprising a first nucleic acid sequence encoding a CEA antigen, a second replication-defective vector comprising a second nucleic acid sequence encoding a Brachyury antigen, a third replication-defective vector comprising a third nucleic acid sequence encoding a MUC1 antigen.

24. The method of claim 23, wherein the CEA antigen is a modified CEA antigen having a least a mutation YLSGANLNL (SEQ ID NO:4) or having an amino acid sequence set forth in YLSGADL L (SEQ ID NO: 10).

25. The method of claim 23 or 24, wherein the first replication-defective vector has a nucleotide sequence at least 80% identical to positions 1057 to 3165 of SEQ ID NO:3.

26. The method of any of claims 23-25, wherein the first replication-defective vector has a nucleotide sequence at least 80% identical to SEQ ID NO:3.

27. The method of any of claims 23-26, wherein the Brachyury antigen binds to HLA-A2.

28. The method of any of claims 23-27, wherein the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence set forth in WLLPGTSTV (SEQ ID NO:22).

29. The method of any of claims 23-28, wherein the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 80% identical to SEQ ID NO:21.

30. The method of any of claims 23-29, wherein the second replication-defective vector has a nucleotide sequence at least 80% identical SEQ ID NO: 8 or positions 1033 to 2283 of SEQ ID NO: 16.

31. The method of any of claims 23-30, wherein the second replication-defective vector has a nucleotide sequence at least 80% identical to SEQ ID NO: 16.

32. The method of any of claims 23-26, wherein the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 80% identical to SEQ ID NO: 15.

33. The method of any of claims 23-26, wherein the second replication-defective vector has a nucleotide sequence at least 80% identical to positions 520-1824 of SEQ ID NO:7.

34. The method of any of claims 23-33, wherein the MUC1 antigen is a modified MUC1 antigen having at least 80% identity to SEQ ID NO:9.

35. The method of any of claims 23-34, wherein the MUC-1 antigen is a modified antigen having one or more mutations at positions 93, 141-142, 149-151, 392, 404, 406, 422, 430-431, 444-445, or 460 of SEQ ID NO:9.

36. The method of any of claims 23-35, wherein the MUC-1 antigen binds to HLA-A2, HLA- A3, HLA-A24, or a combination thereof.

37. The method of any of claims 23-32, wherein the third replication-defective vector has a nucleotide sequence at least 80% identical to SEQ ID NO: 5.

38. The method of any of claims 23-32, wherein the third replication-defective vector has a nucleotide sequence at least 80% identical to SEQ ID NO:6.

39. The method of any of claims 23-32, wherein the third replication-defective vector has a nucleotide sequence at least 80% identical to positions 1033-2858 of SEQ ID NO: 14.

40. The method of any of claims 23-32, wherein the third replication-defective vector has a nucleotide sequence at least 80% identical to SEQ ID NO: 14.

41. The method of claim 23, wherein the CEA antigen is a modified CEA antigen having an amino acid sequence set forth in YLSGADLNL (SEQ ID NO: 10), the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence set forth in WLLPGTSTV (SEQ ID NO:22), and the MUC1 antigen is a modified antigen having one or more mutations at positions 93, 141-142, 149-151, 392, 404, 406, 422, 430-431, 444-445, or 460 of SEQ ID NO:9.

42. A method of treating a CEA-expression cancer in an individual in need thereof, the method comprising:

administering to the individual a first pharmaceutical composition comprising a replication- defective vector comprising a nucleic acid sequence encoding a CEA antigen; and administering to the individual an immune checkpoint inhibitor;

43. A pharmaceutical composition comprising a first replication-defective vector comprising a first nucleic acid sequence encoding a CEA antigen, a second replication-defective vector comprising a second nucleic acid sequence encoding a Brachyury antigen, and a third replication-defective vector comprising a third nucleic acid sequence encoding a MUCl antigen.

44. A method of enhancing an immune response in an individual in need thereof, the method comprising:

administering to the individual a first pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an antigen; and

administering to the individual an immune checkpoint inhibitor.

45. The method of claim 44, wherein the antigen is CEA, Brachyury, MUCl, HPV-E6, HPVE7, or a combination thereof.

46. A method of enhancing an immune response in an individual in need thereof, the method comprising:

administering to the individual a first pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen; and

administering to the individual a chemotherapy, radiation, or a combination thereof.

Description:
METHODS AND COMPOSITIONS FOR COMBINATION IMMUNOTHERAPY

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Application No. 62/150,236, filed April 20, 2015, and is a continuation-in -part of International Application No.

PCT/US2016/012496 filed January 7, 2016, both of which are incorporated herein by reference in their entirety.

SEQUENCE LISTING

[0002] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on April 20, 2016, is named 39891-718-602_SequenceListing.txt and is 208,613 bytes in size.

STATEMENT AS TO FEDERALLY SPONSORED RESEARCH

[0003] This invention was made with government support under Contract No. HHSN

261200900059C, awarded by the National Cancer Institute (NCI); and Contract No. HHSN

261201100097C, awarded by the NCI; Grant No. 1R43CA134063, awarded by the NCI; Grant No. 2R44CA134063 awarded by the NCI; Grant No. 1R43CA186357 awarded by the NCI; Grant No. 1R43DE021973 awarded by the National Institute of Dental and Craniofacial Research (NIDCR); and Grant No. 2R44DE021973 awarded by the NIDCR. The government may have certain rights in the invention.

SUMMARY

[0004] Disclosed herein include methods of enhancing an immune response in an individual in need thereof, the method comprising administering to the individual a first replication-defective vector comprising a first nucleic acid sequence encoding an HPV antigen, and administering to the individual an immune checkpoint inhibitor. In particular embodiments, the antigen used herein is a wild-type HPV antigen or a modified HPV antigen. For example, the modified HPV antigen a non-oncogenic HPV antigen or a modified HPV antigen that has reduced oncogenicity as compared with a wild-type HPV. For example, the antigen used herein is a modified HPV E6 antigen having an amino acid sequence set forth in SEQ ID NO: 17, a modified HPV E7 antigen having an amino acid sequence set forth in SEQ ID NO: 18, or a combination thereof. In further embodiments, the antigen used herein is a modified HPV E6 antigen that has one or more mutations at positions 26, 98, or 106 of SEQ ID NO: 17, a modified HPV E7 antigen that a mutation at position 86 of SEQ ID NO: 18, or a combination thereof. In particular embodiments, the nucleotide sequence of the antigen has a region at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to positions 123-545 of SEQ ID NO: 19, positions 602-895 of SEQ ID NO:9, or a combination thereof. For example, the nucleic acid sequence has at least 80% identity to SEQ ID NO: 19 (the nucleotide sequence of a HPV E6 and E7 fusion protein). In further embodiments, the nucleic acid sequence has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to any portion of or full-length to SEQ ID NO:20 (the predicted sequence of an adenovirus vector expressing HPV E6 and E7), such as positions 1033 to 2179 of SEQ ID NO:20. In certain embodiments, the method may be further defined as treating an HPV infection or an HPV- associated disease, such as an HPV-associated cancer, including, but not limited to, head and neck squamous cell carcinoma (HNSCC), oropharyngeal and tonsillar cancer, anal cancer or cervical cancer.

[0005] In certain embodiments, the method may further comprise administering to the individual an immune checkpoint inhibitor, such as an immune checkpoint inhibitor that targets PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, or CD244. In particular embodiments, the immune checkpoint inhibitor targets PD1 or PDL1, such as anti-PD-1 or anti- PD-L1 antibody, or more particularly, avelumab. In certain embodiments, the method may further comprise administering to the individual chemotherapy, radiation, or a combination thereof. The chemotherapy may be alkylating agents such as cisplatin.

[0006] In certain embodiments, the method is further defined as a method of treating a HPV- associated disease, comprising: administering to the individual a first pharmaceutical

composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen; and administering to the individual an immune checkpoint inhibitor, a chemotherapy, radiation or a combination thereof.

[0007] In certain embodiment, there may be provided a method of enhancing an immune response in an individual in need thereof, the method comprising: administering to the individual a pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen, wherein the administering comprises one or more of the following: i) the administration is repeated at every three weeks; ii) the pharmaceutical composition comprises at least 10 11 adenovirus vectors; iii) the individual is a human; and iv) the individual has , head and neck squamous cell carcinoma (HNSCC), oropharyngeal and tonsillar cancer, anal cancer or cervical cancer.

[0008] In certain embodiments, the replication-defective adenovirus vector has a deletion in an early 2b (E2b) gene region. In further embodiments, the replication-defective adenoviral vector further comprises a deletion in an early 1 (El) gene region, a deletion in an early 3 (E3) gene region, a deletion in an early 4 (E4) gene region, or a combination thereof. In certain embodiments, the CEA, Brachyury or MUC1 antigen has been modified to increase immunogenicity.

[0009] Disclosed herein include methods of enhancing an immune response in an individual in need thereof, the method comprising administering to the individual a first replication-defective vector comprising a first nucleic acid sequence encoding a CEA antigen, a second replication- defective vector comprising a second nucleic acid sequence encoding a Brachyury antigen, a third replication-defective vector comprising a third nucleic acid sequence encoding a MUC1 antigen.

[0010] In certain embodiments, the CEA antigen used herein is a wild-type CEA antigen or a modified CEA antigen having a least a mutation in YLSGANLNL (SEQ ID NO:4), a CAP1 epitope of CEA. The mutation can be conservative or non-conservative, substitution, addition, or deletion. In certain embodiments, the CEA antigen used herein has an amino acid sequence set forth in YLSGADL L (SEQ ID NO: 10), a mutated CAP1 epitope. In further embodiments, the first replication-defective vector or a replication-defective vector that express CEA has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%), 100%) identical to any portion of SEQ ID NO: 3 (the predicted sequence of an adenovirus vector express a modified CEA antigen), such as positions 1057 to 3165 of SEQ ID NO:3 or full-length SEQ ID NO:3.

[0011] In certain embodiments, the Brachyury antigen used herein is a wild-type antigen or a modified antigen. In certain embodiments, the Brachyury antigen binds to HLA-A2. In further embodiments, the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence set forth in WLLPGTSTV (SEQ ID NO:22), a HLA-A2 epitope of Brachyury. In further embodiments, the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%), 100%) identical to SEQ ID NO:21, a modified Brachyury protein sequence. In certain embodiments, the replication-defective vector has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO:8 or positions 1033 to 2283 of SEQ ID NO: 16. In further embodiments, the second replication- defective vector has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to any portion or full-length of SEQ ID NO: 16 (the predicted sequence of an adenovirus vector express a modified Brachyury antigen), such as positions 1033 to 2283 of SEQ ID NO: 16. In some embodiments, the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO: 15 (another mutated Brachyury protein sequence). In certain embodiments, the second replication-defective vector or a replication-defective vector that express Brachyury has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to positions 520-1824 of SEQ ID NO:7 (wild-type Brachyury), SEQ ID NO:7, or SEQ ID NO:8.

[0012] In certain embodiments, the MUCl antigen used herein is a wild-type MUCl antigen or a modified MUCl antigen. In certain embodiments, the modified MUCl antigen has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to SEQ ID NO:9 (a mutated MUCl protein sequence). In certain embodiments, the MUC-1 antigen is a modified antigen having one or more mutations at positions 93, 141-142, 149-151, 392, 404, 406, 422, 430-431, 444-445, or 460 of SEQ ID NO:9. The mutation can be conservative or non-conservative, substitution, addition, or deletion. In further embodiments, the MUC-1 antigen binds to HLA-A2, HLA-A3, HLA-A24, or a combination thereof. In certain embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO:5 (MUC_1 wild-type nucleotide sequence). In further embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO: 6 (a mutated MUCl nucleotide sequence). In certain embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to any portion of or full-length SEQ ID NO: 14 (the predicted sequence of an adenovirus vector express a modified CEA antigen), such as positions 1033-2858 of SEQ ID NO: 14.

[0013] In certain embodiments, there is a provided a composition comprising replication- defective vectors comprising nucleotide sequences encoding a CEA antigen that is a modified CEA antigen having an amino acid sequence set forth in YLSGADLNL (SEQ ID NO: 10), encoding a Brachyury antigen that is a modified Brachyury antigen having an amino acid sequence set forth in WLLPGTSTV (SEQ ID NO:22), and encoding a MUCl antigen that is a modified antigen having one or more mutations at positions 93, 141-142, 149-151, 392, 404, 406, 422, 430-431, 444-445, or 460 of SEQ ID NO:9. In further embodiments, two or more of the first, second, and third, or even more replication-defective vectors are the same vector or in separate vectors.

[0014] In certain embodiments, the method may further comprise administering to the individual an immune checkpoint inhibitor, such as an immune checkpoint inhibitor that targets PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, or CD244. In particular embodiments, the immune checkpoint inhibitor targets PD1 or PDL1, such as anti-PD-1 or anti- PD-L1 antibody, or more particularly, avelumab.

[0015] In further embodiments, the first, second, and third replication-defective vectors are mixed at a 1 : 1 : 1 ratio, or any suitable ratios. In certain embodiments, one or more of the replication-defective vectors disclosed herein, such as the first, second, or third replication- defective vector is a replication-defective adenovirus vector. For example, the replication- defective adenovirus vector has a deletion in an early 2b (E2b) gene region. In further embodiments, the replication-defective adenoviral vector further comprises a deletion in an early 1 (El) gene region, a deletion in an early 3 (E3) gene region, a deletion in an early 4 (E4) gene region, or a combination thereof. In certain embodiments, the CEA, Brachyury or MUC1 antigen has been modified to increase immunogenicity.

[0016] In certain embodiments, methods provided herein may be further defined as treating a cancer that expresses CEA, Brachyury, or MUC1 in an individual in need thereof. In certain embodiments, the cancer to be treated or the individual in need thereof may have breast cancer, lung cancer, head and neck cancer, prostate cancer, gastric cancer, pancreatic cancer, liver cancer, ovarian, cancer, cervical cancer, or gastrointestinal cancer.

[0017] In certain embodiments, any compositions or vectors may be administered to an individual in need thereof sequentially or at the same time. For example, the first, second, or third replication-defective vector is administered to the individual sequentially. In some other embodiments, the first, second, or third replication-defective vector is administered to the individual at the same time.

[0018] In certain embodiments, there is provided a pharmaceutical composition comprising a first replication-defective vector comprising a first nucleic acid sequence encoding a CEA antigen, a second replication-defective vector comprising a second nucleic acid sequence encoding a Brachyury antigen, and a third replication-defective vector comprising a third nucleic acid sequence encoding a MUC1 antigen. Any of the antigens may be wild-type or modified as described as above or throughout the specification.

[0019] Disclosed herein is a method of enhancing an immune response in an individual in need thereof, the method comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an antigen; and administering to the individual an immune checkpoint inhibitor. In certain embodiments, the antigen is CEA, Brachyury, MUC1, an HPV antigen, such as HPV E6 and/or E7, or a combination thereof. In particular embodiments, the antigen used herein is a wild-type HPV antigen or a modified HPV antigen. For example, the modified HPV antigen a non-oncogenic HPV antigen or a modified HPV antigen that has reduced oncogenicity as compared with a wild-type HPV. For example, the antigen used herein is a modified HPV E6 antigen having an amino acid sequence set forth in SEQ ID NO: 17, a modified HPV E7 antigen having an amino acid sequence set forth in SEQ ID NO: 18, or a combination thereof. In further embodiments, the antigen used herein is a modified HPV E6 antigen that has one or more mutations at positions 26, 98, or 106 of SEQ ID NO: 17, a modified HPV E7 antigen that a mutation at position 86 of SEQ ID NO: 18, or a combination thereof. In particular embodiments, the nucleotide sequence of the antigen has a region at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to positions 123-545 of SEQ ID NO: 19, positions 602-895 of SEQ ID NO:9, or a combination thereof. For example, the nucleic acid sequence has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to SEQ ID NO: 19 (the nucleotide sequence of a HPV E6 and E7 fusion protein). In further embodiments, the nucleic acid sequence has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to any portion of or full- length to SEQ ID NO:20 (the predicted sequence of an adenovirus vector expressing HPV E6 and E7), such as positions 1033 to 2179 of SEQ ID NO:20. In certain embodiments, the method may be further defined as treating an HPV infection or an HPV-associated disease, such as an HPV-associated cancer, including, but not limited to, head and neck squamous cell carcinoma (HNSCC), oropharyngeal and tonsillar cancer, anal cancer or cervical cancer.

[0020] In certain embodiments, the method may further comprise administering to the individual an immune checkpoint inhibitor, such as an immune checkpoint inhibitor that targets PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, or CD244. In particular embodiments, the immune checkpoint inhibitor targets PD1 or PDL1, such as anti-PD-1 or anti- PD-L1 antibody, or more particularly, avelumab.

[0021] In certain embodiments, the method may further comprise administering to the individual an immune checkpoint inhibitor, chemotherapy, radiation, or a combination thereof. The chemotherapy may be alkylating agents such as cisplatin. In certain embodiments, the method is further defined as a method of treating a HPV-associated disease, comprising:

administering to the individual a first pharmaceutical composition comprising a replication- defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen; and administering to the individual an immune checkpoint inhibitor, a chemotherapy, radiation or a combination thereof. [0022] In certain embodiments, the replication-defective adenovirus vector has a deletion in an early 2b (E2b) gene region. In further embodiments, the replication-defective adenoviral vector further comprises a deletion in an early 1 (El) gene region, a deletion in an early 3 (E3) gene region, a deletion in an early 4 (E4) gene region, or a combination thereof. In certain

embodiments, the CEA, Brachyury or MUC1 antigen has been modified to increase

immunogenicity.

[0023] In certain embodiments, the vectors have nucleotide sequences encoding one or more of modified CEA, Brachyury or MUC1 antigen. The modified antigens may have insertions, deletions or substitutions, or have been modified to increase immunogenicity. In certain embodiments, the vectors may have nucleotide sequences encoding CEA, Brachyury, MUC1, or a combination thereof. In certain embodiments, two or more antigen-encoding nucleic acids may be comprised in separate vectors or in the same vector.

[0024] In certain embodiments, there is provided a method of treating a CEA-expression cancer in an individual in need thereof, the method comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen or any suitable antigen; and administering to the individual an immune checkpoint inhibitor. The method may further comprise administering to the individual a VEGF inhibitor, chemotherapy, or a combination thereof.

[0025] In certain embodiments, the individual is a human. In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen; and administering to the individual an immune checkpoint inhibitor. In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen; and administering to the individual a chemotherapy. In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication- defective vector comprising a nucleic acid sequence encoding a CEA antigen; administering to the individual an immune checkpoint inhibitor; and administering to the individual a chemotherapy. In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication- defective vector comprising a nucleic acid sequence encoding a CEA antigen; administering to the individual an immune checkpoint inhibitor; administering to the individual a chemotherapy; and administering to the individual a VEGF inhibitor. [0026] In certain embodiments, the CEA antigen is a modified CEA antigen comprising an amino acid sequence set forth in YLSGADLNL (SEQ ID NO: 10). The replication-defective vector comprising a nucleotide sequence encoding a CEA antigen may be provided as described above.

[0027] In certain embodiments, the chemotherapy used herein is capecitabine, leucovorin, fluorouracil, oxaliplatin, fluoropyrimidine, irinotecan, mitomycin, regorafenib, cetuxinab, panitumumab, acetinophen, or a combination thereof. In particular embodiments, the chemotherapy used herein is FOLFOX (leucovorin, fluorouracil and oxaliplatin) or capecitabine. In certain embodiments, the immune checkpoint inhibitor is an anti-PD-1 or anti-PD-Ll antibody, such as avelumab. In certain embodiments, the VEGF inhibitor is an anti-VEGF antibody, such as bevacizumab.

[0028] In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen; administering to the individual an anti-PD-1 or anti-PD-Ll antibody; administering to the individual capecitabine, leucovorin, fluorouracil, oxaliplatin, or a combination thereof; and administering to the individual an anti- VEGF antibody.

[0029] In certain embodiments, the method is further defined as comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen; administering to the individual avelumab; administering to the individual capecitabine or FOLFOX (leucovorin, fluorouracil and oxaliplatin); and administering to the individual bevacizumab.

[0030] In certain embodiments, the method is further defined as comprising: a first treatment phase comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen; administering to the individual avelumab; administering to the individual FOLFOX (leucovorin, fluorouracil and oxaliplatin); and administering to the individual bevacizumab; followed by a second treatment phase, comprising: administered to the individual the first pharmaceutical composition; administering to the individual avelumab; administering to the individual capecitabine; and administering to the individual bevacizumab.

[0031] In certain embodiments, the first pharmaceutical composition, a pharmaceutical composition or a vector disclosed herein is administered intravenously, subcutaneously, intramuscularly or intradermally.

[0032] In certain embodiments, the first pharmaceutical composition, a pharmaceutical composition or a vector disclosed herein is administered every two weeks, every four weeks, or every 12 weeks. In certain embodiments, the first pharmaceutical composition, a pharmaceutical composition or a vector disclosed herein is administered every two weeks, followed by subsequent administration of every four weeks. In certain embodiments, the first pharmaceutical composition, a pharmaceutical composition or a vector disclosed herein is administered every 12 weeks. In certain embodiments, the first pharmaceutical composition, a pharmaceutical composition or a vector disclosed herein is administered every two weeks, followed by subsequent administration of every four weeks, and followed by a second subsequent administration of every 12 weeks.

[0033] In certain embodiments, the first pharmaceutical composition is administered in a treatment phase of up to 24 weeks, wherein the treatment plan further comprises administering to the individual avelumab, FOLFOX, and bevacizumab. In certain embodiments, the first pharmaceutical composition is administered in a treatment phase of up to 24 weeks, wherein the treatment phase further comprises administering to the individual avelumab, FOLFOX, and bevacizumab every two weeks. In certain embodiments, the first pharmaceutical composition comprises at least 10 9 vectors, at least 10 10 vectors or at least 10 11 vectors.

[0034] In certain embodiments, the replication-defective vector is a replication-defective adenovirus vector. In certain embodiments, the replication-defective adenovirus vector has a deletion in an early 2b (E2b) gene region or further comprises a deletion in an early 1 (El) gene region, a deletion in an early 3 (E3) gene region, a deletion in an early 4 (E4) gene region, or a combination thereof.

[0035] In certain embodiments, the first pharmaceutical composition further comprises an immune adjuvant. In certain embodiments, the CEA antigen has been modified to increase immunogenicity.

[0036] In certain embodiments, the first pharmaceutical composition further comprises a vector encoding an antigen selected from the group consisting of Her2/neu, Her3, Brachyury, Brachyury (TIVS7-2, polymorphism), Brachyury (IVS7 T/C polymorphism), T Brachyury, T, PSA, PSCA, PSM (i.e., PSMA), HPV E6, HPV E7, MUC1, MUClc, MUCln, MUC2, p53, BRCA1, MAGE-A1, MAGE-A2, MAGE- A3, MAGE-A4, MAGE-A6, MAGE- A 10, MAGE- A12, BAGE, DAM-6, -10, GAGE-1, -2, -8, GAGE-3, -4, -5, -6, -7B, NA88-A, NY-ESO-1, MART-1, MC1R, GplOO, PAP, Tyrosinase, TRP-1, TRP-2, ART -4, CAMEL, Cyp-B, hTERT, hTRT, iCE, PRAME, P15, RU1, RU2, SART-1, SART-3, WT1, AFP,13-catenin/m, Caspase- 8/m, CDK-4/m, ELF2M, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, Myosin/m, RAGE, SART-2, TRP-2/INT2, 707- AP, Annexin II, CDC27/m, TPLmber- abl, ETV6/AML, LDLR/FUT, Pml/RARa, and TEL/AML1, or a modified tumor antigen or a modified infectious antigen, a splice variant, a functional epitope of a tumor antigen or an infectious antigen, an epitope agonist, or a combination thereof. In particular embodiments, the first pharmaceutical composition further comprises a vector encoding an antigen selected from the group consisting of HPV E6, HPV E7, Her2/neu, MUC1, Her3, Brachyury, PSA, PSM (i.e., PSMA), or a combination thereof.

[0037] In certain embodiments, the pharmaceutical composition is administered before or concurrently with administering to the individual chemotherapy. In certain embodiments, the first pharmaceutical composition is previously frozen.

[0038] In certain embodiments, the individual has previously been administered antihistamine or a pain reliever before administering the immune checkpoint inhibitor. In certain embodiments, the individual has tumor cells that overexpress CEA_as compared with a baseline level in a non- tumor cell. In certain embodiments, the individual has pre-existing immunity to Ad5. In certain embodiments, the individual has cancer of the colon, rectum, breast, lung, pancreas, prostate, gastrointestinal tract, ovary, or a combination thereof. In certain embodiments, the individual has previously been determined to overexpress CEA as compared with a baseline level in a non- tumor cell. In certain embodiments, the individual is or has been determined to have preexisting immunity to adenoviruses.

[0039] In certain embodiments, the pharmaceutical composition further comprises a vector comprising a nucleic acid sequence encoding a costimulatory molecule. In certain embodiments, the replication-defective vector in the first pharmaceutical composition further comprises a nucleic acid sequence encoding a costimulatory molecule. In certain embodiments, the costimulatory molecule comprises B7, ICAM-1, LFA-3, or a combination thereof.

[0040] In certain embodiments, the method is further defined as a method of enhancing an immune response in an individual in need thereof.

INCORPORATION BY REFERENCE

[0041] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

BRIEF DESCRIPTION OF THE DRAWINGS

[0042] FIG. 1 exemplifies a bar graph showing antibody levels from mice immunized with Ad5- null (empty vector). Mice were immunized three times with Ad5-null viral particles (VPs) at 14 day intervals. Anti-Ad antibody (neutralizing antibody) levels increased after each immunization.

[0043] FIG. 2 exemplifies a bar graph showing neutralizing antibody (NAb) levels from mice immunized with Ad5-null. Mice were immunized three times with Ad5-null VPs at 14 day intervals. Neutralizing antibody levels increased after each immunization. Optical density readings indicate the presence of viable target cells.

[0044] FIG. 3 exemplifies a bar graph showing the induction of NAbs in C57B1/6 mice after injections with Ad5-null vector platform. Increasing levels of NAbs were induced in mice after repeated injections with Ad particles.

[0045] FIG. 4A exemplifies a bar graph showing INF-γ levels secreted from splenocytes from Ad5- immune mice immunized with Ad5 [El-]-CEA or Ad5 [E1-, E2b-]-CEA. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA immunized group.

[0046] FIG. 4B exemplifies a bar graph showing IL-2 secreted from splenocytes from Ad5 -immune mice immunized with Ad5 [El-]-CEA or Ad5 [E1-, E2b-]-CEA. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA immunized group.

[0047] FIG. 5 exemplifies a bar graph showing serum aspartate aminotransferase (AST) levels in control mice and mice vaccinated with lxl 0 10 Ad5 [El-]-CEA VPs or Ad5 [E1-, E2b-]-CEA VPs.

[0048] FIG. 6 exemplifies a line graph showing tumor volume over time in Ad5 -immune C57B1/6 mice injected with MC38 CEA-expressing tumor cells and subsequently treated (Vac) with Ad5 [El- , E2b-]-CEA vaccine. Tumor size is shown to be significantly reduced by days 19-21 compared to untreated tumor-bearing mice.

[0049] FIG. 7 exemplifies a graph showing tumor weight from 7 treated and 7 untreated Ad5- immune MC38 tumor-bearing mice. A significant (p=0.0124) reduction in tumor weight in mice treated with Ad5 [E1-, E2b-]-CEA is shown.

[0050] FIG. 8 is an immunoblot using a mouse monoclonal antibody against Gag and exemplifies Gag production by A549 cells infected with Ad5 [E1-, E2b-]-Gag. A549 whole cell lysate was infected with Ad5 [E1-, E2b-]-gag or Ad5-null at a multiplicity of infection (MOI) of 200 for 44 h. The upper band (55kDa) comprises the gag precursor. The lower band (41kDa) comprises the pl7/p24 gag complex.

[0051] FIG. 9 is a graph exemplifying the effect of multiple immunizations on inducing a greater cell-mediated immunity (CMI) response. Ad5-Nai ' ve BALB/C mice (n=5/group) were immunized once or three times at 14 day intervals with lxlO 10 Ad5 [El-]-null VPs, Ad5 [E1-, E2b-]-null VPs, Ad5 [El-]-Gag VPs, Ad5 [E1-, E2b-]-Gag VPs, or buffer alone. IFN-γ secretion from splenocytes was assessed by ELISpot analysis 14 days after the final immunization. Positive control splenocytes were exposed to Concanavalin A (Con A).

[0052] FIG. 10A exemplifies results of an ELISpot INF-γ analysis of peripheral blood mononuclear cells (PBMCs) from Cynomolgus Macaques (n=3) pre-immunized against wild type Ad5. Elevated levels of INF-γ induction (P<0.05) are shown. Positive control splenocytes were exposed to Con A.

[0053] FIG. 10B exemplifies the results of an ELISpot IL-2 analysis from FIG 10A. [0054] FIG. 11 exemplifies a graph depicting the number of spot-forming cells (SFCs) from mice vaccinated with recombinant Ad5 CEA expression vectors secreting IFN-γ. The reduction in SFCs from mice vaccinated with Ad5 [E1-, E2b-]-CEA compared to the reduction in SFCs from Ad5 [El- ]-CEA vaccinated mice is shown.

[0055] FIG. 12A exemplifies a Kaplan-Meier survival plot of 7 patients in cohorts 1 and 2 treated with Ad5 [E1-, E2b-]-CEA(6D).

[0056] FIG. 12B exemplifies a Kaplan-Meier survival plot of 21 patients in cohort 3 and Ph Π treated with Ad5 [E1-, E2b-]-CEA(6D).

[0057] FIG. 12C exemplifies a Kaplan-Meier survival plot of 6 patients in cohort 5 treated with Ad5 [E1-, E2b-]-CEA(6D).

[0058] FIG. 12D exemplifies a Kaplan-Meier survival plot of all 34 patients treated with Ad5 [E1-, E2b-]-CEA(6D).

[0059] FIG. 13A exemplifies a Kaplan-Meier survival plot of 28 cancer patients treated three times with Ad5 [E1-, E2b-]-CEA(6D).

[0060] FIG. 13B exemplifies a Kaplan-Meier survival plot of 27 colorectal cancer patients treated with Ad5 [E1-, E2b-]-CEA(6D).

[0061] FIG. 14 exemplifies CEA-directed CMI responses in treated patients. CMI (IFN-γ secretion) was assessed at baseline (Pre) and after administrations of Ad5 [E1-, E2b-]-CEA(6D) (Post). The highest CMI responses (regardless of time point) observed in the patients after treatment revealed a dose response. The highest CMI levels occurred in patients that received the highest dose of 5x10 11 VP (Cohort 5). The CMI responses for Cohort 3/Phase II and Cohort 5 were significantly elevated (P=0.0002 and P=0.0317, respectively; Mann-Whitney test) as compared to their baseline (Pre) values. Specificity of the responses was demonstrated by the lack of reactivity with the irrelevant antigens β-galactosidase and HTV-gag. For positive controls, PBMCs were exposed to concanavalin A. Values = Mean ± SEM for each Cohort.

[0062] FIG. 15 exemplifies CEA directed CMI responses in treated patients. CMI (IFN-γ secretion) was assessed at baseline (week 0) and 3 weeks after the last immunotherapy (week 9) for patients in all 4-dose cohorts. A dose response is shown and the highest CMI level occurred in patients that received the highest dose. The CMI response with the highest dose was significantly elevated (P<0.02; Mann-Whitney test). Response specificity was shown by the lack of reactivity with the irrelevant antigens β-galactosidase and HTV-gag. Positive control PBMCs were exposed to Con A.

[0063] FIG. 16A exemplifies Ad5 immune responses in patients receiving immunizations with Ad5 [E1-, E2b-]-CEA(6D) vaccine. Ad5 NAb titers to Ad5 were determined in patients at baseline (week 0) and 3 weeks (week 9) after the third immunization. The number of IFN-γ secreting PBMCs from patients specific for Ad5 was determined by ELISpot. The Ad5 NAb titers were significantly elevated at week 9 (pO.0001; Mann-Whitney test). [0064] FIG. 16B exemplifies Ad5 immune responses in patients receiving immunizations with Ad5 [E1-, E2b-]-CEA(6D) vaccine. CMI responses were determined in patients at baseline (week 0) and 3 weeks (week 9) after the third immunization. The number of IFN-γ secreting PBMCs from patients specific for Ad5 was determined by ELISpot. The Ad5 CMI responses were significantly elevated at week 9 (P < 0.01; Mann-Whitney test).

[0065] FIG. 17A exemplifies CEA-specific immunity in patients receiving immunizations with Ad5 [E1-, E2b-]-CEA(6D) vaccine and comparisons with Ad5 immunity. The mean CEA specific immune responses in patients (n=19) who received lxlO 11 VP of Ad5 [E1-, E2b-]-CEA(6D) as measured by IFN-γ secretion of PBMC in patients with none to low pre-existing Ad5 immunity (NAb<200) is shown compared to the CEA specific immune response of patients with high preexisting Ad5 immunity (NAb>200) prior to the initiation of treatment with Ad5 [E1 -, E2b-]- CEA(6D). There was no significant difference between the two groups at any time point tested (p>0.4, Mann-Whitney test)

[0066] FIG. 17B exemplifies a plot of the correlation between pre-existing Ad5 NAb activity and the highest levels of induced CEA CMI responses. The r 2 value (0.0155) indicates there is no correlation between pre-existing Ad5 NAb activity and CEA CMI ELISpot responses.

[0067] FIG. 17C exemplifies a plot of the correlation between vector induced Ad5 NAb activity and CEA CMI responses. The r 2 value (0.0069) indicates there is no correlation between vector- induced Ad5 NAb activity and CEA CMI ELISpot responses.

[0068] FIG. 18 exemplifies a Kaplan-Meier survival plot demonstrating the effect of Ad5 [E1-, E2b-]-CEA(6D) immunotherapy in 32 metastatic colorectal cancer patients (mCRC) patients treated with Ad5 [E1-, E2b-]-CEA(6D).

[0069] FIG. 19 exemplifies CMI responses (IFN-γ secretion) at baseline (Pre-immunized) and after administrations of Ad5 [E1-, E2b-]-CEA(6D) (Post- immunized) in mCRC patients. The highest CMI responses (regardless of time point) observed in the patients after treatment revealed a dose response. The highest CMI levels occurred in patients receiving the highest dose (Cohort 5) and were significantly elevated (p<0.02; Mann-Whitney test). Response specificity is shown by lack of reactivity with irrelevant antigens β-galactosidase and HTV-gag. For positive controls, PBMCs were exposed to Con A.

[0070] FIG. 20 exemplifies CMI responses (ELISpot IFN-γ SFCs) in immunized mCRC patients assessed at weeks 0, 3, 6, and 9. CMI responses increased during immunizations.

[0071] FIG. 21 exemplifies cytotoxic T-cell (CTL) mediated cytotoxicity responses (ELISpot granzyme B secreting SFCs) assessed for pre-immunized (week 0) and post-immunized (weeks 6-9) treatments. Responses increased after immunizations (P < 0.05, Wilcoxon test). [0072] FIG. 22 exemplifies CMI responses in follow-up PBMC samples from 5 immunized mCRC patients as assessed by ELISpot IFN-γ SFCs. CMI responses peaked at week 9 and decreased by week 26 after treatment was stopped.

[0073] FIG. 23 exemplifies CMI responses in mice immunized against CEA, MUCl, and

Brachyury as assessed by ELISpot assays for IFN-γ secretion from splenocytes (IFN-γ SFCs). IFN-γ SFCs were detected in multi -targeted immunized mice (CEA(6D), mMUCl-C, and Brachyury) but not control mice injected with Ad5-Null (empty vector). Response specificity of the ELISpot assay was confirmed by lack of reactivity to irrelevant SIV-nef or SIV-vif peptide antigens. A positive control included cells exposed to Con A.

[0074] FIG. 24A exemplifies a bar graph of results from flow cytometry on polyfunctional CD8a+ splenocyte cells expressing IFN-γ and TNF-a in mice immunized with CEA(6D), mMUCl-C, and Brachyury, but not in controls injected with Ad5-null. Specificity of the responses was confirmed by lack of reactivity to media alone or irrelevant SIV-nef or SIV-vif peptides.

[0075] FIG. 24B exemplifies a bar graph of results from flow cytometry on polyfunctional CD4+ splenocyte cells expressing IFN-γ and TNF-a in mice immunized with CEA(6D), mMUCl-C, and Brachyury, but not in controls injected with Ad5-null. Specificity of the responses was confirmed by lack of reactivity to media alone or irrelevant SIV-nef or SIV-vif peptides.

[0076] FIG. 25A exemplifies a bar graph showing statistical significance (p<0.0001) of CEA antibody-dependent cellular cytotoxicity (ADCC) responses induced in mice immunized with CEA(6D), mMUCl-C, and Brachyury, but not in control mice.

[0077] FIG. 25B exemplifies a bar graph showing statistical significance (p<0.0001) of CEA complement-dependent cellular cytotoxicity (CDCC) responses induced in mice immunized with CEA(6D), mMUCl-C, and Brachyury, but not in control mice.

[0078] FIG. 26A exemplifies results from C57B1/6 mice (n=7/group) subcutaneously inoculated in the left flank with MC38 tumor cells expressing MUCl tumor cells and administered lxlO 10 Ad5- null VPs or lxlO 10 Ad5 [E1-, E2b-]-mMUCl-C VPs in the right flank on days 0, 7, 14. Mice treated with Ad5 [E1-, E2b-]-mMUCl-C had significantly (p<0.05) smaller tumors on days 15 and 18 compared to controls and significantly longer survival. Experiments were terminated on day 36.

[0079] FIG. 26B exemplifies that the mice treated with Ad5 [E1-, E2b-]-mMUCl-C as described for Fig. 26A had significantly (p<0.05) longer survival on days 15 and 18 compared to controls.

[0080] FIG. 27 exemplifies results from C57B1/6 mice subcutaneously inoculated in the left flank with MC38 tumor cells expressing Brachyury and administered lxlO 10 Ad5-null VPs (n=4) or lxlO 10 Ad5 [E1-, E2b-] -Brachyury VPs (n=5) in the right flank on days 5, 11, and 17. Tumors were smaller in treated mice on days 15, 19, and 22.

[0081] FIG. 28 exemplifies the effects of HPV immunotherapy in C57B1/6 mice (n=7/group) implanted with HPV-E6/E7 expressing TC-1 tumor cells (day 0) and treated by immunotherapy on days 10, 17, 24 with lxlO 10 Ad5-null VPs plus 100 μg control IgG (intraperitoneal), lxlO 10 Ad5-null VPs plus 100 μg anti-PDl, lxl 0 10 Ad5 [E1-, E2b-]-HPV-E6/E7 VPs plus 100 μg mouse IgG, or lxlO 10 Ad5 [E1-, E2b-]-HPV-E6/E7 VPs plus 100 μg anti-PDl. Immunotherapy with or without anti-PDl resulted in significant inhibition of tumor growth by day 23 (p<0.05). All control mice were terminated by day 23 due to tumor mass.

[0082] FIG. 29 exemplifies combination multi -targeted HPV immunotherapy effects on tumor growth and regression in 2 mice treated by immunotherapy with anti-PDl injections in FIG. 28.

Tumor growth peaked on days 20 and 27, respectively, and then regressed thereafter.

[0083] FIG. 30A exemplifies expression of Brachyury protein in human dendritic cells (DCs) infected with Ad5 [E1-, E2b-] -Brachyury. SW620 tumor cells were used as positive control. Actin was used as a loading control. Expression of Brachyury was robust in DCs infected with Ad5 [E1 -,

E2b-]-Brachyury.

[0084] FIG. 30B exemplifies expression of MUC1 protein in human DCs infected with Ad5 [E1-, E2b-]-MUCl. SW620 tumor cells were used as positive control. Actin was used as a loading control. MUC1 expression was observed in human DCs infected with Ad5 [E1-, E2b-]-MUCl vector as compared to DCs infected with Ad5 [E1-, E2b-]-null.

[0085] FIG. 31A exemplifies analyses of IFN-γ secreted from splenocytes from C57B1/6 mice (n = 5/group) vaccinated three times at 2-week intervals with lxlO 10 Ad5 [E1-, E2b-] -Brachyury VPs, lxl0 10 Ad5 [E1-, E2b-]-CEA VPs, lxl0 10 Ad5 [E1-, E2b-]-MUCl VPs, or Tn-Ad5 (1 : 1 : 1 mixture of lxlO 10 Ad5 [E1-, E2b-] -Brachyury VPs, lxlO 10 Ad5 [E1-, E2b-]-CEA VPs, and lxlO 10 Ad5 [E1-, E2b-]-MUCl VPs). Controls received 3xl0 10 Ad5-null VPs. Splenocytes were collected 14 days after the final vaccination and assessed for IFN-γ secretion by ELISpot assay. Positive control splenocytes were exposed to Con A. Significant differences (p<0.05) between columns are reported in p-values. Not significant = ns.

[0086] FIG. 31B exemplifies analyses of IL-2 secreted from splenocytes from the vaccinated mice described in FIG. 31A. Splenocytes were assessed for IL-2 secretion by ELISpot assay.

[0087] FIG. 32A exemplifies a graph of analyses of CD8 + and multifunctional cellular populations following vaccination of C57B1/6 mice (n=5/group) vaccinated three times at 2-week intervals with lxl0 10 Ad5 [E1-, E2b-] -Brachyury VPs, lxl0 10 Ad5 [E1-, E2b-]-CEA VPs, lxl0 10 Ad5 [E1-, E2b-]- MUC1 VPs or Tn-Ad5 (1 : 1 : 1 mixture of lxlO 10 Ad5 [E1-, E2b-] -Brachyury VPs, lxl0 10 Ad5 [E1-, E2b-]-CEA VPs, and lxl0 10 Ad5 [E1-, E2b-]-MUCl VPs). Controls received 3xl0 10 Ad5 [E1-, E2b- ]-null VPs. Splenocytes were collected 14 days after the final vaccination and were assessed by FACS for CD8a + cells secreting IFN-γ and TNF-a. Positive control splenocytes were exposed to Con A.

[0088] FIG. 32B exemplifies a graph of FACS analyses of CD4 + cells and multifunctional cellular populations from the vaccinated mice described in FIG. 32A. [0089] FIG. 32C exemplifies a graph of FACS analyses of CD8a + cells secreting IFN-γ and TNF-a from the vaccinated mice described in FIG. 32A.

[0090] FIG. 32D exemplifies a graph of FACS analyses of CD4 + cells secreting IFN-γ and TNF-a from the vaccinated mice described in FIG. 32A.

[0091] FIG. 33A exemplifies an ELISA analysis of CEA IgG levels in mice vaccinated three times with lxlO 10 Ad5 [E1-, E2b-]-CEA VPs, Tn-Ad5 (1 : 1 : 1 mixture of lxlO 10 Ad5 [E1-, E2b-]- Brachyury VPs, lxl0 10 Ad5 [E1-, E2b-]-CEA VPs, and lxl0 10 Ad5 [E1-, E2b-]-MUCl VPs), or 3x10 10 Ad5 [E1-, E2b-]-null VPs.

[0092] FIG. 33B exemplifies a CDC assay against MC38-CEA2 cells using the mice described in FIG. 33A

[0093] FIG. 34 exemplifies effects of immunotherapy on tumor volume in C57B1/6 mice

(n=7/group) inoculated subcutaneously in the left flank with lxlO 6 MC-38-MUC1 cells and lxlO 10 Ad5 [E1-, E2b-]-MUCl VPs or Tn-Ad5 (1 : 1: 1 mixture of lxl0 10 Ad5 [E1-, E2b-]-CEA VPs, lxl0 10 Ad5 [E1-, E2b-]-MUCl VPs, and lxlO 10 Ad5 [E1-, E2b-]-Brachyury VPs (3xl0 10 VP total)). Control mice received 3xl0 10 Ad5 [E1-, E2b-]-null VPs. (*) indicates days when Ad5 [E1-, E2b-]- MUC1 treated mice had significantly smaller (p<0.05) tumors than control mice and ( A ) indicates days when Tri-Ad5-treated mice had significantly smaller (p<0.05) tumors than control mice. No significant difference (p>0.1) between Ad5 [E1-, E2b-]-MUCl and Tri-Ad5-treated mice was seen.

[0094] FIG. 35 exemplifies an immunoblot showing expression of CEA in A549 cells infected with Ad5 [E1-, E2b-]-CEA. (A) Negative Control. (B) Protein molecular weight marker. (C) Negative. (D) CEA Reference Material (30 ng). (E) Ad5 [E1-, E2b-]-CEA lysate (20 μΕ). (F) Ad5 [E1-, E2b-]- CEA lysate (20 μΕ). (G) Negative A549 cells. Recombinant CEA was used as a positive control and uninfected A549 cells served as a negative control.

[0095] FIG. 36 exemplifies CMI dose responses as measured by ELISpot of splenocytes from C57BL/6 mice (n=5/group) immunized three times at 14-day intervals with doses of lxlO 8 , lxlO 9 or lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs and assessed 14 days after the final immunization. The greatest induction of CMI was achieved with the lxl 0 10 VP dose. Positive control splenocytes were exposed to Con A.

[0096] FIG. 37A exemplifies activation of CD8-a7lFN-Y + splenocytes after immunization of C57BL/6 mice (n=5/group) immunized three times at two week intervals with lxlO 10 VP Ad5 [E1-, E2b-]-E6/E7 VPs. Controls received lxlO 10 Ad5 [E1-, E2b-]-null VPs. Splenocytes collected 14 days after the final immunization were assessed by flow cytometry for. For positive controls, splenocytes were exposed to PMA/ionomycin.

[0097] FIG. 37B exemplifies activation of CD8-a + /IFN-Y + /TNF-a + splenocytes after immunization of mice as described in FIG. 37A. [0098] FIG. 38A exemplifies changes in tumor size from immunotherapy of C57BL/6 mice (n=5/group) implanted on day 0 with 2x10 5 non-palpable HFV-E6/E7 TC-1 tumor cells and administered lxlO 10 Ad5 [E1-, E2b-]-null VPs or lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs on days 1, 8 and 15. Tumor size was determined and volumes calculated according to the formula V = (a 2 x b)/2. Analysis of significance was performed between experimental and vector control groups using unpaired t-tests and significance is denoted by * (p<0.05) and ** (p<0.01).

[0099] FIG. 38B exemplifies a survival curve of the mice as described in FIG. 38A that was plotted and compared using the Mantel-Cox test. Significance is denoted by ** (p<0.01).

[00100] FIG. 39A exemplifies changes in tumor size from immunotherapy of C57BL/6 mice (n=4/group) implanted on day 0 with 2x10 5 small palpable HPV-E6/E7 TC-1 tumor cells and administered lxlO 10 Ad5 [E1-, E2b-]-null VPs or lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs on days 6, 13 and 20. Tumor size was determined and volumes calculated according to the formula V = (a 2 x b)/2. Analysis of significance was performed between experimental and vector control groups using unpaired t-tests and significance is denoted by ** (p<0.01).

[00101] FIG. 39B exemplifies a survival curve of the mice as described in FIG. 39A that was plotted and compared using the Mantel-Cox test. Significance is denoted by ** (p<0.01).

[00102] FIG. 40A exemplifies changes in tumor size from immunotherapy of C57BL/6 mice (n=4/group) implanted on day 0 with 2x10 5 large established HPV-E6/E7 TC-1 tumor cells and administered lxlO 10 Ad5 [E1-, E2b-]-null VPs or lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs on days 13, 20 and 27. Tumor size was determined and volumes calculated according to the formula V = (a 2 x b)/2. Analysis of significance was performed between experimental and vector control groups using unpaired t-tests and significance is denoted by ** (p<0.01).

[00103] FIG. 40B exemplifies a survival curve of the mice as described in FIG. 40A that was plotted and compared using the Mantel-Cox test. Significance is denoted by ** (p<0.01).

[00104] FIG. 41A exemplifies changes in tumor size from C57BL/6 mice (n=7/group) inoculated on day 0 with 2x10 5 TC-1 tumor cells and administered treatments on days 10, 17, and 24 with lxl 0 10 Ad5 [E1-, E2b-]-null VPs plus 100 μg of isotype control rat IgG. Tumor size was determined and volumes calculated according to the formula V = (a 2 x b)/2. Tumor growth kinetics represents individual mice in each group.

[00105] FIG. 41B exemplifies changes in tumor size from C57BL/6 mice (n=7/group) inoculated on day 0 with 2x10 5 TC-1 tumor cells and administered treatments on days 10, 17, and 24 with lxlO 10 Ad5 [E1-, E2b-]-null VPs plus 100 μg anti-PDl antibody.

[00106] FIG. 41C exemplifies changes in tumor size from C57BL/6 mice (n=7/group) inoculated on day 0 with 2x10 5 TC-1 tumor cells and administered treatments on days 10, 17, and 24 with lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs plus 100 μg isotype control rat IgG. [00107] FIG. 41D exemplifies changes in tumor size from C57BL/6 mice (n=7/group) inoculated on day 0 with 2x10 5 TC-1 tumor cells and administered treatments on days 10, 17, and 24 with lxlO 10 Ad5 [E1-, E2b-]-E6/E7 VPs plus 100 μg anti-PDl.

[00108] FIG. 42 exemplifies a survival curve for C57BL/6 mice (n=7/group) treated as those in FIGs. 41A-D. The experiment was terminated on day 52 following tumor implantation. Mice treated with Ad5 [E1-, E2b-]-E6/E7 and control antibody exhibited significantly (p < 0.008) longer survival compared to both groups of control mice (Ad5 [E1-, E2b-]-null and control antibody or Ad5 [E1-, E2b-]-null and anti-PDl antibody). 2 of 7 (29%) Ad5 [E1-, E2b-]-E6/E7 and control antibody treated mice remained alive at day 52. Mice treated with Ad5 [E1 -, E2b-]-E6/E7 plus anti PD1 antibody exhibited significantly (p < 0.0006) longer survival as compared to both groups of controls. 4 of 7 (57%) Ad5 [E1-, E2b-]-E6/E7 plus anti-PDl antibody treated mice remained alive at day 52.

[00109] FIG. 43A exemplifies that Ad5 [El -, E2b-]-E6/E7 promotes the recruitment of CD8+ tumor-infiltrating lymphocytes (TILs) into TC-1 tumors. C57BL/6 mice (n=5/group) were implanted with 2x105 TC-1 tumor cells. Twelve days after implantation mice began treatment with Ad5 [E1 -, E2b-]-null empty vector plus control IgG, Ad5 [E1-, E2b-]-null plus anti-PDl, Ad5 [E1-, E2b-]- E6/E7 plus control IgG, or Ad5 [E1-, E2b-]-E6/E7 plus anti-P-1. Vaccine was administered subcutaneously weekly and anti-PDl antibodies were administered via intraparietal injection every 3-4 days and tumors were analyzed on day 27. Ad5 [E1-, E2b-]-E6/E7 treatment significantly decreases the ratio of Treg/CD8+ TILs. Analysis of significance was performed using unpaired t- tests and significance is denoted by ns (p>0.05), * (p<0.05), ** (pO.01), *** (pO.001), or **** (pO.0001).

[00110] FIG. 43B exemplifies that the reduction in the ratio of Treg/CD8+ TILs of FIG. 43A reduction is not driven by a reduction in the number of Tregs.

[00111] FIG. 43C exemplifies that the reduction in the ratio of Treg/CD8+ TILs of FIG. 43A is driven through an increase in the number of CD8+ TILs.

[00112] FIG.44A exemplifies that Ad5 [E1-, E2b-]-E6/E7 plus anti-PDl antibody combination therapy promotes a pro -inflammatory tumor microenvironment. C57BL/6 mice (n=5/group) were tumor implanted, treated, and tumors were analyzed as in FIGs. 43A-C. The frequency of PD1 + CD4 + and CD8 + TILs is increased in tumors from mice treated with Ad5 [E1-, E2b-]-E6/E7. Tumors from mice treated with a combination of Ad5 [E1-, E2b-]-E6/E7 and anti-PDl have a significantly lower frequency of PD1 + CD4 + and CD8 + TILs (A), LAG-3 + CD8 + TILs (B), and (C). Analysis of significance was performed using unpaired t-tests and significance is denoted by ns (p>0.05), * (p<0.05), ** (pO.01), or *** (pO.001).

[00113] FIG.44B exemplifies that tumors from mice treated with a combination of Ad5 [E1-, E2b- ]-E6/E7 and anti-PDl as in FIG. 44A have a significantly lower frequency of LAG-3 + CD8 + TILs bringing these levels more in line with tumors from control mice. [00114] FIG.44C exemplifies that tumors from mice treated with a combination of Ad5 [E1-, E2b- ]-E6/E7 and anti-PDl as in FIG. 44A have a significantly reduced expression level of PDL1.

[00115] FIG. 45A exemplifies a bar graph showing INF-γ levels secreted from splenocytes from Ad5 -naive mice immunized with Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). Splenocytes were also assessed for non-specific INF-γ secreting T-cells by stimulation with the non-immunizing antigens β-galactosidase (β-gal) and HTV-1 Gag. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA(6D) immunized group. The errors bars depict the SEM.

[00116] FIG. 45B exemplifies a bar graph showing IL-2 levels secreted from splenocytes from Ad5 -naive mice immunized with Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). Splenocytes were also assessed for non-specific IL-2 secreting T-cells by stimulation with the non-immunizing antigens β-galactosidase (β-gal) and HTV-1 Gag. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA(6D) immunized group. The errors bars depict the SEM.

[00117] FIG. 46A exemplifies a bar graph showing INF-γ levels secreted from splenocytes from Ad5 -immune mice immunized with Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). Splenocytes were also assessed for non-specific INF-γ secreting T-cells by stimulation with the non-immunizing antigens β-galactosidase (β-gal) and HTV-1 Gag. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA(6D) immunized group. The errors bars depict the SEM.

[00118] FIG. 46B exemplifies a bar graph showing IL-2 levels secreted from splenocytes from Ad5 -immune mice immunized with Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). Splenocytes were also assessed for non-specific IL-2 secreting T-cells by stimulation with the non-immunizing antigens β-galactosidase (β-gal) and HTV-1 Gag. A significantly elevated response is shown in the Ad5 [E1-, E2b-]-CEA(6D) immunized group. The errors bars depict the SEM.

[00119] FIG. 47 exemplifies a bar graph showing INF-γ levels secreted from splenocytes from CEA transgenic mice immunized with Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone

(control). Elevated levels of IFN-γ secreting splenocytes specific for CEA and not for control antigens such as CMV or HTV-1 Gag is shown. The error bars depict the SEM.

[00120] FIG. 48A exemplifies a line graph showing tumor volume over time in Ad5-nai ' ve C57B1/6 mice injected with MC-38 CEA-expressing tumor cells and subsequently treated with Ad5 [E1-, E2b-]-CEA vaccine. Tumor size is shown to be significantly reduced by days 20-21 compared to untreated tumor-bearing mice. * Indicates a significant difference in the value of the means on that day. Values represent Mean ± SEM. [00121] FIG. 48B exemplifies a graph showing tumor weight from 7 treated and 7 untreated Ad5- naive MC38 tumor-bearing mice. A significant (p<0.05) reduction in tumor weight in mice treated with Ad5 [E1-, E2b-]-CEA is shown. Values represent Mean ± SEM.

[00122] FIG. 48C exemplifies a bar graph showing INF-γ levels secreted from splenocytes from Ad5-naive mice immunized with Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). A significantly (p<0.05) elevated response is shown in the Ad5 [E1 -, E2b-]-CEA(6D) immunized group. Values represent Mean ± SEM.

[00123] FIG. 49A exemplifies a line graph showing tumor volume over time in Ad5-immune C57B1/6 mice injected with MC-38 CEA-expressing tumor cells and subsequently treated with Ad5 [E1-, E2b-]-CEA vaccine. Tumor size is shown to be significantly reduced by days 19-21 compared to untreated tumor-bearing mice. * Indicates a significant difference in the value of the means on that day. Values represent Mean ± SEM.

[00124] FIG. 49B exemplifies a graph showing tumor weight from 7 treated and 7 untreated Ad5- immune MC38 tumor-bearing mice. A significant (p<0.05) reduction in tumor weight in mice treated with Ad5 [E1-, E2b-]-CEA is shown. Values represent Mean ± SEM.

[00125] FIG. 49C exemplifies a bar graph showing INF-γ levels secreted from splenocytes from Ad5-immune mice immunized with Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control). A significantly (p<0.05) elevated response is shown in the Ad5 [E1 -, E2b-]-CEA(6D) immunized group. Values represent Mean ± SEM.

[00126] FIG. 50 exemplifies bar graphs showing profiles of CEA directed CMI responses in 5 mCRC patients during and after the course of immunotherapy. A decrease in CMI response from their peak values after immunizations ended was shown. Values are Mean ± SEM.

[00127] FIG. 51A exemplifies Kaplan-Meier survival plot on long-term overall survival of treated mCRC patients. Panel represents all treated patients. There were 23 events during the study.

[00128] FIG. 51B exemplifies Kaplan-Meier survival plot on long-term overall survival of treated mCRC patients. Panel represents patients that received all 3 treatments. There were 23 events during the study.

[00129] FIG. 51C exemplifies Kaplan-Meier survival plot on long-term overall survival of treated mCRC patients. Panel represents patients immunized 3 times with the 2 highest doses of vaccine. There were 23 events during the study.

[00130] FIG.52 exemplifies immunotherapy combined with chemotherapy/ radiation treatment of established HPV-E6/E7 expressing tumors. Established HPV-E6/E7 expressing tumors were treated by immunotherapy on days 7, 14, & 21 combined with cisplatin/radiation treatment on days 13, 20, & 27. Control tumor bearing mice were treated by injections with Ad -null combined with cisplatin/radiation treatment. [00131] FIG.53 exemplifies bar graph for CMI responses in mice treated with combination therapy. . Non-tumor bearing mice were treated as described in FIG. 44. Two weeks after the last treatment, mice were assessed for CMI activity as determined by ELISpot assays for IFN-γ secreting splenocytes. Increased CMI responses in mice treated with combination therapy (Ad5 [E1 -, E2b-]- mE6/E7/CRT) was shown.

[00132] FIG. 54 exemplifies clinical outcomes for colorectal cancer patients with avelumab by RECIST criteria.

[00133] FIG. 55 exemplifies CEA change in patient with mCRC receiving single agent avelumab (MSB0010718C).

[00134] FIG. 56 exemplifies MC 38 Tumor Growth Inhibition in Mice Receiving Avelumab (Anti- PD-L1) in Combination with Oxaliplatin and 5-FU. Female C57BL/6 mice were inoculated in the right subcutaneous flank with 1x106 MC38 colon carcinoma cells. When tumors reached a mean volume of -50 mm3, mice were sorted into treatment groups (N=10). 5-FU (60 mg/kg by i.v. injection) and OX (5 mg/kg by i.p. injection) were administered on days 0 and 14.

MSB0010718C (400 μg by i.v.injection) was given on days 3, 6, and 9. Tumor volumes were measured twice weekly via calipers. Data are mean ± SEM.

[00135] FIG. 57 exemplifies survival of MC38 Tumor-Bearing Mice Receiving Avelumab (Anti- PD-L1) in Combination with Oxaliplatin and 5-FU. Female C57BL/6 mice were inoculated in the right subcutaneous flank with lxl 0 6 MC38 colon carcinoma cells. When tumors reached a mean volume of -50 mm3, mice were sorted into treatment groups (N=10). 5-FU (60 mg/kg by i.v.

injection) and OX (5 mg/kg by i.p. injection) were administered on days 0 and 14. MSB0010718C (400 μg by i.v. injection) was given on days 3, 6, and 9. Kaplan-Meier plots and Log Rank statistics were used to calculate survival differences between groups. Survival was censored at the time tumor volumes reached 2000 mm3. Data are expressed as median survival time in days.

[00136] FIG. 58 exemplifies Pentamer Analysis of Tumor- Antigen Specific T Cells. Subsets of 5 animals from each study group were sacrificed on study day 21 for FACS-based detection of pl5Especific T cells using a fluorescently-labeled synthetic MHC class I pentamer loaded with a pl5E-derived antigenic peptide epitope. The spleen from each animal was dissociated into single cell suspensions and subsequently stained with an anti-CD8 antibody and the pl5E pentamer. From the total splenocyte gate, the percentage of cells positive for both CD8 and the pentamer was determined. Data were acquired using a BD LSR II flow cytometer.

[00137] FIG. 59 exemplifies Splenic CD8+/PD-1+ T cell Counts. Subsets of 5 animals from each group were sacrificed for FACS-based immunophenotyping on day 21 after the start of treatment. Spleens were harvested and dissociated into single cell suspensions by mechanical disruption.

Suspensions were then stained with fluorescently-labeled antibodies specific for relevant T cell surface markers (i.e.. CD8, PD-1, CD44, CD62L) and data were acquired on a BD LSR II flow cytometer.

[00138] FIG. 60 exemplifies CD8+ Tumor-Infiltrating Lymphocyte Levels. Subsets of 5 animals from each group were sacrificed for FACS-based immunophenotyping analysis on day 21 following the start of treatment. Tumors were harvested and dissociated into single cell suspensions by enzymatic digestion (collagenase) and mechanical disruption. Suspensions were then stained with a fluorescently-labeled antibodies specific for the NKl. l surface marker and data were acquired using a BD LSR II flow cytometer.

[00139] FIG. 61 exemplifies Tumor-Infiltrating NK Cell Levels. Subsets of 5 animals from each group were sacrificed for FACS-based immunophenotyping analysis on day 21 following the start of treatment. Tumors were harvested and dissociated into single cell suspensions by enzymatic digestion (collagenase) and mechanical disruption. Suspensions were then stained with a

fluorescently-labeled antibodies specific for the NKl. l surface marker and data were acquired using a BD LSR II flow cytometer.

[00140] FIG. 62 exemplifies Combination of Avelumab (MSB0010718C) with 5-FU and

Oxaliplatin: ELISPOT Measurement of Tumor Antigen Specific CD8+ T cells. For the ELISPOT assay, spleens from treated animals were harvested on day 21 after the start of treatment and processed into single cells suspensions (splenocytes from 5 mice from each group were pooled). The splenocytes were cultured for six days in the presence of a pl5E-derived peptide antigen epitope (amino acid sequence KSPWFTTL) in order to stimulate expansion of antigen-specific CD8+ T cells. As a control for non-specific activation, the assay was run in parallel using an irrelevant peptide derived from chicken ovalbumin (OVA). Following the six day period of in vitro antigen stimulation, the CD8+ T cells were isolated from the cultures and placed in co-culture with antigen presenting cells (APCs) loaded with the same pl5E antigenic peptide or OVA as a control. The cocultures were prepared in ELISPOT assay plates coated with an antibody that captured secreted IFN-γ. Following assay plate development, each IFN-γ positive "spot" represented a CD8+ T cell that had become activated by pl5E-specific stimulation.

[00141] FIG. 63 exemplifies Assessment and Initial Management of Tumor Lysis Syndrome (TLS).

[00142] FIG. 64A exemplifies adverse effects in Ad5 naive mice following immunization with Ad5 [El ]-CEA(6D) or Ad5 [E1-, E2b-]-CEA(6D). Adverse effects were determined by evaluation of the liver enzyme aspartate aminotransferase (AST) levels in serum from C57B1/6 mice

(n=7/group) immunized at a weekly interval with 10 10 VP of Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]- CEA(6D) or injection buffer alone. Three days after one immunization (Day 3) or three

immunizations (Day 17), AST levels were determined. The error bars depict the SEM. [00143] FIGS. 64A-64B exemplifies adverse effects in Ad5 naive mice following immunization with Ad5 [El-]-CEA(6D) or Ad5 [E1-, E2b-]-CEA(6D). Adverse effects were determined by evaluation of the liver enzyme alanine aminotransferase (ALT) levels in serum from C57B1/6 mice (n=7/group) immunized at a weekly interval with 10 10 VP of Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]- CEA(6D) or injection buffer alone. Three days after one immunization (Day 3) or three

immunizations (Day 17), ALT levels were determined. The error bars depict the SEM.

[00144] FIGS. 65A-65B exemplifies adverse effects in Ad5 immune mice following immunization with Ad5 [El-]-CEA(6D) or Ad5 [E1-, E2b-]-CEA(6D). Adverse effects were determined by evaluation of the liver enzyme aspartate aminotransferase (AST) levels in serum from Ad5 immune C57B1/6 mice immunized three times at a weekly interval with 10 10 VP of Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D) or injection buffer alone. Three days after three immunizations (Day 17), AST levels were determined. The error bars depict the SEM.

[00145] FIG. 66 exemplifies a randomization schema of the clinical trial (the star indicates that patients on standard of care are allowed to cross-over to Avelumab + Ad-CEA vaccine + standard of care at progression of disease).

[00146] FIG. 67 exemplifies a dosing regimen of the clinical trial.

[00147] FIG. 68 exemplifies CMI response as assessed by flow cytometry. C57BL/6 mice were immunized three times with 10 10 VP Ad5[El-,E2b-]-null or 10 10 VP Ad5[El-,E2b-]-E6/E7 at two week intervals. Two weeks after the final immunization CD8a+ splenocytes were assayed for intracellular expression of IFNy after 6 hour stimulation with antigen specific peptide pools. Mean +/- standard deviation is plotted.

[00148] FIG. 69 exemplifies a result of immunotherapy of small established HPV-E6/E7 expressing tumors with Ad5 [E1-, E2b-]-E6/E7. C57BL/6 mice were implanted on day 0 with 2xl0 5 TC-1 tumor cells and administered 10 10 VP Ad5 [E1-, E2b-]-null (vector control) or 10 10 VP Ad5 [E1-, E2b-]-E6/E7 on days 6, 13 and 20 as indicated by arrows. (A) Tumor size was determined and volumes calculated according to the formula V=(a 2 xb)/2. On day 23, mice were euthanized from the vector control group. No analyses of significance could be performed after this 23 day time point and this is denoted by a dashed line. Analysis of significance was performed between experimental and vector control groups using unpaired t-tests and significance is denoted by ** (p<0.01). Error bars represent the standard error of the means.

DETAILED DESCRIPTION

[00149] The following passages describe different aspects of certain embodiments in greater detail. Each aspect may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature of features indicated as being preferred or advantageous. [00150] Unless otherwise indicated, any embodiment can be combined with any other embodiment. A variety of aspects can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range as if explicitly written out. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range. When ranges are present, the ranges include the range endpoints.

[00151] Compared to first generation adenovirus vectors, certain embodiments of the Second Generation E2b deleted adenovirus vectors contain additional deletions in the DNA polymerase gene (pol) and deletions of the pre-terminal protein (pTP). E2b deleted vectors have up to a 13 kb gene- carrying capacity as compared to the 5 to 6 kb capacity of First Generation adenovirus vectors, easily providing space for nucleic acid sequences encoding any of a variety of target antigens. The E2b deleted adenovirus vectors also have reduced adverse reactions as compared to first generation adenovirus vectors.

[00152] The innate immune response to wild type Ad can be complex, and it appears that Ad proteins expressed from adenovirus vectors play an important role. Specifically, the deletions of preterminal protein and DNA polymerase in the E2b deleted vectors appear to reduce inflammation during the first 24 to 72 h following injection, whereas First Generation adenovirus vectors stimulate inflammation during this period. In addition, it has been reported that the additional replication block created by E2b deletion also leads to a 10,000 fold reduction in expression of Ad late genes, well beyond that afforded by El, E3 deletions alone. The decreased levels of Ad proteins produced by E2b deleted adenovirus vectors effectively reduce the potential for competitive, undesired, immune responses to Ad antigens, responses that prevent repeated use of the platform in Ad immunized or exposed individuals. The reduced induction of inflammatory response by second generation E2b deleted vectors results in increased potential for the vectors to express desired vaccine antigens during the infection of antigen presenting cells (i.e., dendritic cells), decreasing the potential for antigenic competition, resulting in greater immunization of the vaccine to the desired antigen relative to identical attempts with First Generation adenovirus vectors. E2b deleted adenovirus vectors provide an improved Ad-based vaccine candidate that is safer, more effective, and more versatile than previously described vaccine candidates using First Generation adenovirus vectors.

Thus, first generation, El -deleted Adenovirus subtype 5 (Ad5)-based vectors, although promising platforms for use as cancer vaccines, are impeded in activity by naturally occurring or induced Ad- specific neutralizing antibodies. Without being bound by theory, Ad5 -based vectors with deletions of the El and the E2b regions (Ad5 [E1-, E2b-]), the latter encoding the DNA polymerase and the preterminal protein, for example by virtue of diminished late phase viral protein expression, may avoid immunological clearance and induce more potent immune responses against the encoded tumor antigen transgene in Ad-immune hosts.

[00153] Some embodiments relate to methods and compositions (e.g., viral vectors) for generating immune responses against target antigens, in particular, those associated or related to infectious disease or proliferative cell disease such as cancer. Some embodiments relate to methods and compositions for generating immune responses in an individual against target antigens, in particular, those related to cell proliferation diseases such as cancer. In some embodiments, compositions and methods described herein relate to generating an immune response in an individual against cells expressing and/or presenting a target antigen or a target antigen signature comprising at least one target antigen. Some embodiments provide compositions and methods for immunotherapy against human papilloma virus (HPV) using a viral gene delivery platform to immunize against HPV genes E6 and E7 combined with PD1 checkpoint blockade. These compositions and methods utilize an Ad5 [E1-, E2b-]-E6/E7 vaccine combined with an immune pathway checkpoint modulator.

[00154] The compositions and methods can be used to generate an immune response against a target antigen expressed and/or presented by a cell. For example, the compositions and methods can be used to generate immune responses against a carcinoembryonic antigen (CEA), such as CEA expressed or presented by a cell. For example, the compositions and methods can be used to generate an immune response against CEA(6D) expressed or presented by a cell. For example, the compositions and methods can be used to generate an immune response against Mucin 1 (MUCl) expressed and/or presented by a cell. For example, the compositions and methods can be used to generate an immune response against MUClc expressed and/or presented by a cell. For example, the compositions and methods can be used to generate an immune response against Brachyury (T protein (T)) expressed and/or presented by a cell.

[00155] The compositions and methods can be used to generate an immune response against multiple target antigens expressed and/or presented by a cell. For example, the compositions and methods can be used to generate an immune response against MUClc, T, or any combination thereof. For example, the compositions and methods can be used to generate an immune response against T and CEA. For example, the compositions and methods can be used to generate an immune response against MUClc and CEA. For example, the compositions and methods can be used to generate an immune response against MUCl and T. For example, the compositions and methods can be used to generate an immune response against MUClc, T, and CEA.

[00156] A modified form of CEA, MUClc, or T can be used in a vaccine directed to raising an immune response against CEA, MUClc, or T, or cells expressing and/or presenting CEA, MUClc, or T. In particular, some embodiments provide an improved Ad-based vaccine such that multiple vaccinations against one or more antigenic target entity can be achieved. In some embodiments, the improved Ad-based vaccine comprises a replication defective adenovirus carrying a target antigen, a fragment, a variant or a variant fragment thereof, such as Ad5 [E1-, E2b-]-CEA(6D). Variants or fragments of target antigens, such as CEA, MUClc, or T, can be selected based on a variety of factors, including immunogenic potential. A mutant CEA, CEA(6D) can utilized for its increased capability to raise an immune response relative to the CEA(WT). Importantly, vaccination can be performed in the presence of preexisting immunity to the Ad or administered to subjects previously immunized multiple times with the Ad vector as described herein or other Ad vectors. The Ad vectors can be administered to subjects multiple times to induce an immune response against an antigen of interest, such as CEA, MUClc, or T, including but not limited to, the production of antibodies and CMI responses against one or more target antigens.

[00157] As used herein, unless otherwise indicated, the article "a" means one or more unless explicitly otherwise provided for. As used herein, unless otherwise indicated, terms such as "contain," "containing," "include," "including," and the like mean "comprising." As used herein, unless otherwise indicated, the term "or" can be conjunctive or disjunctive. As used herein, unless otherwise indicated, any embodiment can be combined with any other embodiment.

[00158] An "adenovirus" (Ad) refers to non-enveloped DNA viruses from the family

Adenoviridae. These viruses can be found in, but are not limited to, human, avian, bovine, porcine and canine species. Some embodiments contemplate the use of any Ad from any of the four genera of the family Adenoviridae (e.g., Aviadeno virus, Mastadeno virus, Atadeno virus and Si adenovirus) as the basis of an E2b deleted virus vector, or vector containing other deletions as described herein. In addition, several serotypes are found in each species. Ad also pertains to genetic derivatives of any of these viral serotypes, including but not limited to, genetic mutations, deletions or

transpositions.

[00159] A "helper adenovirus" or "helper virus" refers to an Ad that can supply viral functions that a particular host cell cannot (the host may provide Ad gene products such as El proteins). This virus is used to supply, in trans, functions (e.g., proteins) that are lacking in a second virus, or helper dependent virus (e.g., a gutted or gutless virus, or a virus deleted for a particular region such as E2b or other region as described herein); the first replication-incompetent virus is said to "help" the second, helper dependent virus thereby permitting the production of the second viral genome in a cell.

[00160] An "adenovirus 5 null (Ad5-null)" refers to a non-replicating Ad that does not contain any heterologous nucleic acid sequences for expression.

[00161] A "first generation adenovirus" refers to an Ad that has the early region 1 (El) deleted. In additional cases, the early region 3 (E3) may also be deleted. [00162] "Gutted" or "gutless" refers to an Ad vector that has been deleted of all viral coding regions.

[00163] "Transfection" refers to the introduction of foreign nucleic acid into eukaryotic cells. Exemplary means of transfection include calcium phosphate-DNA co-precipitation, DEAE-dextran- mediated transfection, polybrene-mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, retroviral infection, and biolistics.

[00164] "Stable transfection" or "stably transfected" refers to the introduction and integration of foreign nucleic acid, DNA or RNA, into the genome of the transfected cell. The term "stable transfectant" refers to a cell which has stably integrated foreign DNA into the genomic DNA.

[00165] A "reporter gene" indicates a nucleotide sequence that encodes a reporter molecule (e.g., an enzyme). A "reporter molecule" is detectable in any of a variety of detection systems, including, but not limited to, enzyme-based detection assays (e.g., ELISA, histochemical assays), fluorescent, radioactive, and luminescent systems. The E. coli β-galactosidase gene, green fluorescent protein (GFP), the human placental alkaline phosphatase gene, the chloramphenicol acetyltransferase (CAT) gene; and other reporter genes may be employed.

[00166] A "heterologous sequence" refers to a nucleotide sequence that is ligated to, or is manipulated to become ligated to, a nucleic acid sequence to which it is not ligated in nature, or to which it is ligated at a different location in nature. Heterologous nucleic acid may include a naturally occurring nucleotide sequence or some modification relative to the naturally occurring sequence.

[00167] A "transgene" refers to any gene coding region, either natural or heterologous nucleic acid sequences or fused homologous or heterologous nucleic acid sequences, introduced into cells or a genome of subject. Transgenes may be carried on any viral vector used to introduce transgenes to the cells of the subject.

[00168] A "second generation adenovirus" refers to an Ad that has all or parts of the El, E2, E3, and, in certain embodiments, E4 DNA gene sequences deleted (removed) from the virus.

[00169] A "subject" refers to any animal, including, but not limited to, humans, non-human primates (e.g., rhesus or other types of macaques), mice, pigs, horses, donkeys, cows, sheep, rats and fowls.

[00170] An "immunogenic fragment" refers to a fragment of a polypeptide that is specifically recognized (i.e., specifically bound) by a B-cell and/or T-cell surface antigen receptor resulting in a generation of an immune response specifically against a fragment.

[00171] A "target antigen" or "target protein" refers to a molecule, such as a protein, against which an immune response is to be directed.

[00172] "E2b deleted" refers to a DNA sequence mutated in such a way so as to prevent expression and/or function of at least one E2b gene product. Thus, in certain embodiments, "E2b deleted" is used in relation to a specific DNA sequence that is deleted (removed) from an Ad genome. E2b deleted or "containing a deletion within an E2b region" refers to a deletion of at least one base pair within an E2b region of an Ad genome. Thus, in certain embodiments, more than one base pair is deleted and in further embodiments, at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 base pairs are deleted. In another embodiment, a deletion is of more than 150, 160, 170, 180, 190, 200, 250, or 300 base pairs within an E2b region of an Ad genome. An E2b deletion may be a deletion that prevents expression and/or function of at least one E2b gene product and therefore, encompasses deletions within exons of encoding portions of E2b-specific proteins as well as deletions within promoter and leader sequences. In certain embodiments, an E2b deletion is a deletion that prevents expression and/or function of one or both a DNA polymerase and a preterminal protein of an E2b region. In a further embodiment, "E2b deleted" refers to one or more point mutations in a DNA sequence of this region of an Ad genome such that one or more encoded proteins is non-functional. Such mutations include residues that are replaced with a different residue leading to a change in an amino acid sequence that result in a nonfunctional protein.

[00173] "El -deleted" refers to a DNA sequence that is mutated in such a way so as to prevent expression and/or function of at least one El gene product. Thus, in certain embodiments, "El deleted" is used in relation to a specific DNA sequence that is deleted (removed) from the Ad genome. El deleted or "containing a deletion within the El region" refers to a deletion of at least one base pair within the El region of the Ad genome. Thus, in certain embodiments, more than one base pair is deleted and in further embodiments, at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 base pairs are deleted. In another embodiment, the deletion is of more than 150, 160, 170, 180, 190, 200, 250, or 300 base pairs within the El region of the Ad genome. An El deletion may be a deletion that prevents expression and/or function of at least one El gene product and therefore, encompasses deletions within exons of encoding portions of El -specific proteins as well as deletions within promoter and leader sequences. In certain embodiments, an El deletion is a deletion that prevents expression and/or function of one or both of a trans-acting transcriptional regulatory factor of the El region. In a further embodiment, "El deleted" refers to one or more point mutations in the DNA sequence of this region of an Ad genome such that one or more encoded proteins is non-functional. Such mutations include residues that are replaced with a different residue leading to a change in the amino acid sequence that result in a nonfunctional protein.

[00174] "Generating an immune response" or "inducing an immune response" refers to a statistically significant change, e.g., increase or decrease, in the number of one or more immune cells (T-cells, B-cells, antigen-presenting cells, dendritic cells, neutrophils, and the like) or in the activity of one or more of these immune cells (CTL activity, HTL activity, cytokine secretion, change in profile of cytokine secretion, etc.).

[00175] The terms "nucleic acid" and "polynucleotide" are used essentially interchangeably herein. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be DNA (e.g. genomic, cDNA, or synthetic) or RNA molecules. RNA molecules may include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide as described herein, and a polynucleotide may, but need not, be linked to other molecules and/or support materials. An isolated polynucleotide, as used herein, means that a polynucleotide is substantially away from other coding sequences. For example, an isolated DNA molecule as used herein does not contain large portions of unrelated coding DNA, such as large chromosomal fragments or other functional genes or polypeptide coding regions. This refers to the DNA molecule as originally isolated, and does not exclude genes or coding regions later added to the segment recombinantly in the laboratory.

[00176] As will be understood by those skilled in the art, the polynucleotides can include genomic sequences, extra-genomic and plasmid-encoded sequences and smaller engineered gene segments that express, or may be adapted to express target antigens as described herein, fragments of antigens, peptides and the like. Such segments may be naturally isolated, or modified synthetically by the hand of man.

[00177] Typically, polynucleotide variants will contain one or more substitutions, additions, deletions and/or insertions, preferably such that the immunogenicity of the epitope of the polypeptide encoded by the variant polynucleotide or such that the immunogenicity of the heterologous target protein is not substantially diminished relative to a polypeptide encoded by the native polynucleotide sequence. In some cases, the one or more substitutions, additions, deletions and/or insertions may result in an increased immunogenicity of the epitope of the polypeptide encoded by the variant polynucleotide. As described elsewhere herein, the polynucleotide variants can encode a variant of the target antigen, or a fragment (e.g., an epitope) thereof wherein the propensity of the variant polypeptide or fragment (e.g., epitope) thereof to react with antigen-specific antisera and/or T-cell lines or clones is not substantially diminished relative to the native

polypeptide. The polynucleotide variants can encode a variant of the target antigen, or a fragment thereof wherein the propensity of the variant polypeptide or fragment thereof to react with antigen- specific antisera and/or T-cell lines or clones is substantially increased relative to the native polypeptide.

[00178] The term "variants" should also be understood to encompass homologous genes of xenogenic origin. In particular embodiments, variants or fragments of target antigens are modified such that they have one or more reduced biological activities. For example, an oncogenic protein target antigen may be modified to reduce or eliminate the oncogenic activity of the protein, or a viral protein may be modified to reduce or eliminate one or more activities or the viral protein. An example of a modified CEA protein is a CEA having a N610D mutation, resulting in a variant protein with increased immunogenicity. [00179] When comparing polynucleotide sequences, two sequences are "identical" if the sequence of nucleotides in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software using default parameters.

Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman, Add. APL. Math 2:482 (1981), by the identity alignment algorithm of Needleman and Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity methods of Pearson and Lipman, Proc. Natl. Acad. Sci. USA 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA), or by inspection. One example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms. BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. In one illustrative example, cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLASTN program uses as defaults a word length (W) of 11, and expectation (E) of 10, and the BLOSUM62 scoring matrix alignments, (B) of 50, expectation (E) of 10, M=5, N=-4 and a comparison of both strands.

[00180] The "percentage of sequence identity" can be determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence and multiplying the results by 100 to yield the percentage of sequence identity.

[00181] It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a particular antigen of interest, or fragment thereof, as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of some embodiments. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).

Viral Vectors for Immunotherapies and Vaccines

[00182] Recombinant viral vectors can be used to express protein coding genes or antigens (e.g., TAAs (tumor-associated antigens) and/or IDAAs (infectious-disease associated antigens)). The advantages of recombinant viral vector based vaccines and immunotherapy include high efficiency gene transduction, highly specific delivery of genes to target cells, induction of robust immune responses, and increased cellular immunity. Certain embodiments provide for recombinant adenovirus vectors comprising deletions or insertions of crucial regions of the viral genome. The viral vectors of provided herein can comprise heterologous nucleic acid sequences that encode one or more target antigens of interest, or variants, fragments or fusions thereof, against which it is desired to generate an immune response.

[00183] Suitable viral vectors that can be used with the methods and compositions as provided herein, include but are not limited to retroviruses, lentiviruses, provirus, Vaccinia virus,

adenoviruses, adeno-associated viruses, self-complementary adeno-associated virus,

Cytomegalovirus, or Sendai virus. In some embodiments, the viral vector can be replication- competent. In some embodiments, the viral vector can be replication-defective. For replication- defective viral vectors, the viruses' genome can have the coding regions necessary for additional rounds of replication and packaging replaced with other genes, or deleted. These viruses are capable of infecting their target cells and delivering their viral payload, but then fail to continue the typical lytic pathway that leads to cell lysis and death. Depending on the viral vector, the typical maximum length of an allowable DNA or cDNA insert in a replication-defective viral vector is can be about 8- lO kilobases (kB).

[00184] Retroviruses have been used to express antigens, such as an enveloped, single -stranded RNA virus that contains reverse transcriptase. Retrovirus vectors can be replication-defective.

Retrovirus vectors can be of murine or avian origin. Retrovirus vectors can be from Moloney murine leukemia virus (MoMLV). Retrovirus vectors can be used that require genome integration for gene expression. Retrovirus vectors can be used to provide long-term gene expression. For example, retrovirus vectors can have a genome size of approximately 7-11 kb and the vector can harbor 7-8 kb long foreign DNA inserts. Retrovirus vectors can be used to display low immunogenicity and most patients do not show pre-existing immunity to retroviral vectors. Retrovirus vectors can be used to infect dividing cells. Retrovirus vectors can be used to not infect non-dividing cells.

[00185] Lentivirus vectors have been used to express antigens. Lentiviruses constitute a subclass of retroviruses. Lentivirus vectors can be used to infect non-dividing cells. Lentivirus vectors can be used to infect dividing cells. Lentivirus vectors can be used to infect both non-dividing and dividing cells. Lentiviruses generally exhibit broader tropism than retroviruses. Several proteins such as tat and rev regulate the replication of lentiviruses. These regulatory proteins are typically absent in retroviruses. HTV is an exemplary lentivirus that can been engineered into a transgene delivery vector. The advantages of lentivirus vectors are similar to those of retroviral vectors. Although lentiviruses can potentially trigger tumorigenesis, the risk is lower than that of retroviral vectors, as the integration sites of lentiviruses are away from the sites harboring cellular promoters. HTV -based vectors can be generated, for example, by deleting the HTV viral envelope and some of the regulatory genes not required during vector production. Instead of parental envelope, several chimeric or modified envelope vectors are generated because it determines the cell and tissue specificity.

[00186] Cytomegalovirus (CMV) vectors have been used to express antigens and is a member of the herpesviruses. Species-specific CMVs can be used (e.g., human CMV (HCMV), e.g., human herpesvirus type 5. HCMV contains a 235-kb double-stranded linear DNA genome surrounded by a capsid. The envelope contains glycoproteins gB and gH, which bind to cellular receptors.

[00187] Sendai virus (SeV) vectors have been used to express antigens. SeV is an enveloped, single-stranded RNA virus of the family Paramyxovirus. The SeV genome encodes six protein and two envelope glycoproteins, HN and F proteins, that mediate cell entry and determine its tropism. SeV vectors that lack F protein can be used as a replication-defective virus to improve the safety of the vector. SeV vector produced in a packaging cell can be used to expresses the F protein. An F gene-deleted and transgene-inserted genome can be transfected into a packaging cell. SeV contains RNA dependent RNA polymerase and viral genome localizes to the cytoplasm. This ensures that fast gene expression occurs soon after infection and the genotoxic advantage of SeV. SeV vectors can be used to exhibit highly efficient gene transfer. SeV vectors can be used to transduce both dividing and non-dividing cells. SeV vectors can be used to transduce non-dividing cells. SeV vectors can be used to transduce dividing cells. SeV vectors can be used, for example, to efficiently transduce human airway epithelial cells. SeV vectors can be, for example, administered by a mucosal (e.g., oral and nasal) route. Intranasal administration can be used to potentially reduce the influence of a pre- existing immunity to SeV, as compared to intramuscular administration. Compared to other viral vectors, its transgene capacity (3.4 kb) is low. SeV is highly homologous to the human parainfluenza type 1 (hPrV-1) virus; thus, a pre-existing immunity against hPIV-1 can work against the use of SeV.

[00188] Human papillomavirus (HFV) vectors can be used to express antigens. For example, by modifying oncogenes in the genome, such as by deletion or insertion of crucial regions of the HFV viral genome, a recombinant vector can be engineered to increase predictability of infection and reduce unwanted side effects. An exemplary HPV vector is a fusion vector with an adenovirus vector. An exemplary HPV vector is Ad5 [E1-, E2b-]-HPV-E6/E7 viral vector comprising a modified non-oncogenic and fused HPV-E6/E7.

Adenovirus Vectors

[00189] In general, adenoviruses are attractive for clinical because they can have a broad tropism, they can infect a variety of dividing and non-dividing cell types and hey can be used systemically as well as through more selective mucosal surfaces in a mammalian body. In addition, their relative thermostability further facilitates their clinical use. Adenoviruses are a family of DNA viruses characterized by an icosahedral, non-enveloped capsid containing a linear double-stranded genome. Generally, adenoviruses are found as non-enveloped viruses comprising double-stranded DNA genome approximated -30-35 kilobases in size. Of the human Ads, none are associated with any neoplastic disease, and only cause relatively mild, self-limiting illness in immunocompetent individuals. The first genes expressed by the virus are the El genes, which act to initiate high-level gene expression from the other Ad5 gene promoters present in the wild type genome. Viral DNA replication and assembly of progeny virions occur within the nucleus of infected cells, and the entire life cycle takes about 36 hr with an output of approximately 10 4 virions per cell. The wild type Ad5 genome is approximately 36 kb, and encodes genes that are divided into early and late viral functions, depending on whether they are expressed before or after DNA replication. The early/late delineation is nearly absolute, since it has been demonstrated that super-infection of cells previously infected with an Ad5 results in lack of late gene expression from the super-infecting virus until after it has replicated its own genome. Without bound by theory, this is likely due to a replication dependent cw-activation of the Ad5 major late promoter (MLP), preventing late gene expression (primarily the Ad5 capsid proteins) until replicated genomes are present to be encapsulated. The composition and methods as described herein, in some embodiments, take advantage of feature in the development of advanced generation Ad vectors/vaccines. The linear genome of the adenovirus is generally flanked by two origins for DNA replication (ITRs) and has eight units for RNA polymerase II-mediated transcription. The genome carries five early units El A, E1B, E2, E3, E4, and E5, two units that are expressed with a delay after initiation of viral replication (IX and IVa2), and one late unit (L) that is subdivided into L1-L5. Some adenoviruses can further encode one or two species of RNA called virus-associated (VA) RNA.

[00190] Adenoviruses that induce innate and adaptive immune responses in human patient are provided. By deletion or insertion of crucial regions of the viral genome, recombinant vectors are provided that have been engineered to increase their predictability and reduce unwanted side effects. In some aspects, there is provided an adenovirus vector comprising the genome deletion or insertion selected from the group consisting of: El A, E1B, E2, E3, E4, E5, IX, IVa2, LI, L2, L3, L4, and L5, and any combination thereof.

[00191] Certain embodiments provide recombinant adenovirus vectors comprising an altered capsid. Generally, the capsid of an adenovirus is primarily comprises 20 triangular facets of an icosahedron each icosahedron contains 12 copies of hexon trimers. In addition there are also other several additional minor capsid proteins, Ilia, VI, VIII, and IX.

[00192] Certain embodiments provide recombinant adenovirus vectors comprising one or more altered fiber proteins. In general the fiber proteins, which also form trimers, are inserted at the 12 vertices into the pentameric penton bases. The fiber can comprise of a thin N-terminal tail, a shaft, and a knob domain. The shaft can comprise a variable numbers of β-strand repeats. The knob can comprise one or more loops A, B, C, D, E, F, G, H, I, J. The fiber knob loops can bind to cellular receptors. Certain embodiments provide adenovirus vectors to be used in vaccine systems for the treatment of cancers and infectious diseases.

[00193] Suitable adenoviruses that can be used with the present methods and compositions of the disclosure include but are not limited to species-specific adenovirus including human subgroups A, Bl, B2, C, D, E and F or their crucial genomic regions as provided herein, which subgroups can further classified into immunologically distinct serotypes. Further, suitable adenoviruses that can be used with the present methods and compositions of the disclosure include, but are not limited to, species-specific adenovirus or their crucial genomic regions identified from primates, bovines, fowls, reptiles, or frogs.

[00194] Some adenoviruses serotypes preferentially target distinct organs. Serotypes such as AdHul, AdHu2, and AdHu5 (subgenus C), generally effect the infect upper respiratory, while subgenera A and F effect gastrointestinal organs. Certain embodiments provide recombinant adenovirus vectors to be used in preferentially target distinct organs for the treatment of organ- specific cancers or organ-specific infectious diseases. In some applications the recombinant adenovirus vector is altered to reduce tropism to a specific organ in a mammal. In some applications the recombinant adenovirus vector is altered to increase tropism to a specific organ in a mammal.

[00195] The tropism of an adenovirus can be determined by their ability to attach to host cell receptors. In some instances the process of host cell attachment can involve the initial binding of the distal knob domain of the fiber to a host cell surface molecule followed by binding of the RGD motif within the penton base with aV integrins. Certain embodiments provide recombinant adenovirus vectors with altered tropism such that they can be genetic engineered to infect specific cell types of a host. Certain embodiments provide recombinant adenovirus vectors with altered tropism for the treatment of cell-specific cancers or cell-specific infectious diseases. Certain embodiments provide recombinant adenovirus vectors with altered fiber knob from one or more adenoviruses of subgroups A, B, C, D, or F, or a combination thereof or the insertion of RGD sequences. In some applications the recombinant adenovirus vectors comprising an altered fiber knob results in a vector with reduced tropism for one or more particular cell types. In some applications the recombinant adenovirus vectors comprising an altered fiber knob results in a vector with enhanced tropism for one or more particular cell types. In some applications the recombinant adenovirus vectors comprising an altered fiber knob results in a vector with reduced product-specific B or T-cell responses. In some applications the recombinant adenovirus vectors comprising an altered fiber knob results in a vector with enhanced product-specific B or T-cell responses.

[00196] Certain embodiments provide recombinant adenovirus vectors that are coated with other molecules to circumvent the effects of virus-neutralizing antibodies or improve transduction in to a host cell. Certain embodiments provide recombinant adenovirus vectors that are coated with an adaptor molecule that aids in the attachment of the vector to a host cell receptor. By way of example an adenovirus vector can be coated with adaptor molecule that connects coxsackie Ad receptor (CAR) with CD40L resulting in increased transduction of dendritic cells, thereby enhancing immune responses in a subject. Other adenovirus vectors similarly engineered for enhancing the attachment to other target cell types are also contemplated.

Ad5 Vectors

[00197] Studies in humans and animals have demonstrated that pre-existing immunity against Ad5 can be an inhibitory factor to commercial use of Ad-based vaccines. The preponderance of humans have antibody against Ad5, the most widely used subtype for human vaccines, with two-thirds of humans studied having lympho -proliferative responses against Ad5. This pre-existing immunity can inhibit immunization or re-immunization using typical Ad5 vaccines and may preclude the immunization of a vaccine against a second antigen, using an Ad5 vector, at a later time.

Overcoming the problem of pre-existing anti -vector immunity has been a subject of intense investigation. Investigations using alternative human (non-Ad5 based) Ad5 subtypes or even non- human forms of Ad5 have been examined. Even if these approaches succeed in an initial immunization, subsequent vaccinations may be problematic due to immune responses to the novel Ad5 subtype. To avoid the Ad5 immunization barrier, and improve upon the limited efficacy of first generation Ad5 [E1-] vectors to induce optimal immune responses, some embodiments relate to a next generation Ad5 vector based vaccine platform. [00198] First generation, or El -deleted adenovirus vectors Ad5 [E1-] are constructed such that a transgene replaces only the El region of genes. Typically, about 90% of the wild-type Ad5 genome is retained in the vector. Ad5 [E1-] vectors have a decreased ability to replicate and cannot produce infectious virus after infection of cells that do not express the Ad5 El genes. The recombinant Ad5 [E1-] vectors are propagated in human cells (e.g., 293 cells) allowing for Ad5 [E1-] vector replication and packaging. Ad5 [E1-] vectors have a number of positive attributes; one of the most important is their relative ease for scale up and cGMP production. Currently, well over 220 human clinical trials utilize Ad5 [E1-] vectors, with more than two thousand subjects given the virus sc, im, or iv. Additionally, Ad5 vectors do not integrate; their genomes remain episomal. Generally, for vectors that do not integrate into the host genome, the risk for insertional mutagenesis and/or germ- line transmission is extremely low if at all. Conventional Ad5 [E1-] vectors have a carrying capacity that approaches 7 kb.

[00199] Ad5-based vectors with deletions of the El and the E2b regions (Ad5 [El -, E2b-]), the latter encoding the DNA polymerase and the pre-terminal protein, by virtue of diminished late phase viral protein expression, provide an opportunity to avoid immunological clearance and induce more potent immune responses against the encoded tumor antigen transgene in Ad-immune hosts. The new Ad5 platform has additional deletions in the E2b region, removing the DNA polymerase and the preterminal protein genes. The Ad5 [E1-, E2b-] platform has an expanded cloning capacity that is sufficient to allow inclusion of many possible genes. Ad5 [E1-, E2b-] vectors have up to about 12 kb gene-carrying capacity as compared to the 7 kb capacity of Ad5 [E1 -] vectors, providing space for multiple genes if needed. In some embodiments, an insert of more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 kb is introduced into an Ad5 vector, such as the Ad5 [E1-, E2b-] vector. Deletion of the E2b region confers advantageous immune properties on the Ad5 vectors, often eliciting potent immune responses to target transgene antigens while minimizing the immune responses to Ad viral proteins.

[00200] In various embodiments, Ad5 [E1-, E2b-] vectors induce a potent CMI, as well as antibodies against the vector expressed vaccine antigens even in the presence of Ad immunity. Ad5 [E1-, E2b-] vectors also have reduced adverse reactions as compared to Ad5 [E1-] vectors, in particular the appearance of hepatotoxicity and tissue damage. A key aspect of these Ad5 vectors is that expression of Ad late genes is greatly reduced. For example, production of the capsid fiber proteins could be detected in vivo for Ad5 [E1-] vectors, while fiber expression was ablated from Ad5 [E1-, E2b-] vector vaccines. The innate immune response to wild type Ad is complex. Proteins deleted from the Ad5 [E1-, E2b-] vectors generally play an important role. Specifically, Ad5 [E1-, E2b-] vectors with deletions of preterminal protein or DNA polymerase display reduced

inflammation during the first 24 to 72 h following injection compared to Ad5 [E1-] vectors. In various embodiments, the lack of Ad5 gene expression renders infected cells invisible to anti-Ad activity and permits infected cells to express the transgene for extended periods of time, which develops immunity to the target.

[00201] Some embodiments contemplate increasing the capability for the Ad5

[E1-, E2b-] vectors to transduce dendritic cells, improving antigen specific immune responses in the vaccine by taking advantage of the reduced inflammatory response against Ad5 [E1 -, E2b-] vector viral proteins and the resulting evasion of pre-existing Ad immunity.

Replication defective Ad5 vector

[00202] Attempts to overcome anti-Ad immunity have included use of alternative Ad serotypes and/or alternations in the Ad5 viral capsid protein each with limited success and the potential for significantly altering biodistribution of the resultant vaccines. Therefore, a completely novel approach was attempted by further reducing the expression of viral proteins from the El deleted Ad5 vectors, proteins known to be targets of pre-existing Ad immunity. Specifically, a novel recombinant Ad5 platform has been described with deletions in the early 1 (El) gene region and additional deletions in the early 2b (E2b) gene region (Ad5 [E1-, E2b-]). Deletion of the E2b region (that encodes DNA polymerase and the pre-terminal protein) results in decreased viral DNA replication and late phase viral protein expression. This vector platform can be used to induce CMI responses in animal models of cancer and infectious disease and more importantly, this recombinant Ad5 gene delivery platform overcomes the barrier of Ad5 immunity and can be used in the setting of preexisting and/or vector-induced Ad immunity thus enabling multiple homologous administrations of the vaccine. In particular embodiments, some embodiments relate to a replication defective adenovirus vector of serotype 5 comprising a sequence encoding an immunogenic polypeptide. The immunogenic polypeptide may be a mutant, natural variant, or a fragment thereof.

[00203] In some embodiments, the replication defective adenovirus vector comprises a modified sequence encoding a polypeptide with at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to a wild-type immunogenic polypeptide or a fragment thereof. In some embodiments, the replication defective adenovirus vector comprises a modified sequence encoding a subunit of a wild-type polypeptide. The compositions and methods, in some embodiments, relate to an adenovirus-derived vector comprising at least 60% sequence identity to SEQ. ID. NO:3.

[00204] In some embodiments, an adenovirus-derived vector, optionally relating to a replication defective adenovirus, comprises a sequence with at least 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.8%, 99.9% identity to SEQ. ID. NO:3 or a sequence generated from SEQ. ID. NO:3 by alternative codon replacements. In various embodiments, the adenovirus-derived vectors described herein have a deletion in the E2b region, and optionally, in the El region, the deletion conferring a variety of advantages to the use of the vectors in immunotherapy as described herein. [00205] Certain regions within the adenovirus genome serve essential functions and may need to be substantially conserved when constructing the replication defective adenovirus vectors. These regions are further described in Lauer et al, J. Gen. Virol, 85, 2615-25 (2004), Leza et al, J. Virol, p. 3003-13 (1988), and Miralles et al, J. Bio Chem., Vol. 264, No. 18, p. 10763-72 (1983), which are incorporated by reference in their entirety. Recombinant nucleic acid vectors comprising a sequence with identity values of at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.8%, 99.9%, 100% to a portion of SEQ. ID. NO: 3, such as a portion comprising at least about 100, 250, 500, 1000 or more bases of SEQ. ID. NO: 3 are used in some embodiments.

[00206] Certain embodiments contemplate the use of E2b deleted adenovirus vectors, such as those described in U.S. Pat. Nos. 6,063,622; 6,451,596; 6,057,158; 6,083,750; and 8,298,549, which are each incorporated herein by reference in their entirety. The vectors with deletions in the E2b regions in many cases cripple viral protein expression and/or decrease the frequency of generating replication competent Ad (RCA). Propagation of these E2b deleted adenovirus vectors can be done utilizing cell lines that express the deleted E2b gene products. Such packaging cell lines are provided herein; e.g., E.C7 (formally called C-7), derived from the HEK-2p3 cell line.

[00207] Further, the E2b gene products, DNA polymerase and preterminal protein, can be constitutively expressed in E.C7, or similar cells along with the El gene products. Transfer of gene segments from the Ad genome to the production cell line has immediate benefits: (1) increased carrying capacity; and, (2) a decreased potential of RCA generation, typically requiring two or more independent recombination events to generate RCA. The El, Ad DNA polymerase and/or preterminal protein expressing cell lines used in some embodiments can enable the propagation of adenovirus vectors with a carrying capacity approaching 13 kb, without the need for a contaminating helper virus. In addition, when genes critical to the viral life cycle are deleted (e.g., the E2b genes), a further crippling of Ad to replicate or express other viral gene proteins occurs. This can decrease immune recognition of infected cells, and extend durations of foreign transgene expression.

[00208] El, DNA polymerase, and preterminal protein deleted vectors are typically unable to express the respective proteins from the El and E2b regions. Further, they may show a lack of expression of most of the viral structural proteins. For example, the major late promoter (MLP) of Ad is responsible for transcription of the late structural proteins LI through L5. Though the MLP is minimally active prior to Ad genome replication, the highly toxic Ad late genes are primarily transcribed and translated from the mLP only after viral genome replication has occurred. This cis- dependent activation of late gene transcription is a feature of DNA viruses in general, such as in the growth of polyoma and SV-40. The DNA polymerase and preterminal proteins are important for Ad replication (unlike the E4 or protein IX proteins). Their deletion can be extremely detrimental to adenovirus vector late gene expression, and the toxic effects of that expression in cells such as APCs. [00209] The adenovirus vectors can include a deletion in the E2b region of the Ad genome and, optionally, the El region. In some cases, such vectors do not have any other regions of the Ad genome deleted. The adenovirus vectors can include a deletion in the E2b region of the Ad genome and deletions in the El and E3 regions. In some cases, such vectors have no other regions deleted. The adenovirus vectors can include a deletion in the E2b region of the Ad genome and deletions in the El, E3 and partial or complete removal of the E4 regions. In some cases, such vectors have no other deletions. The adenovirus vectors can include a deletion in the E2b region of the Ad genome and deletions in the El and/or E4 regions. In some cases, such vectors contain no other deletions. The adenovirus vectors can include a deletion in the E2a, E2b and/or E4 regions of the Ad genome. In some cases, such vectors have no other deletions. The adenovirus vectors can have the El and/or DNA polymerase functions of the E2b region deleted. In some cases, such vectors have no other deletions. The adenovirus vectors can have the El and/or the preterminal protein functions of the E2b region deleted. In some cases, such vectors have no other deletions. The adenovirus vectors can have the El, DNA polymerase and/or the preterminal protein functions deleted. In some cases, such vectors have no other deletions. The adenovirus vectors can have at least a portion of the E2b region and/or the El region. In some cases, such vectors are not gutted adenovirus vectors. In this regard, the vectors may be deleted for both the DNA polymerase and the preterminal protein functions of the E2b region. The adenovirus vectors can have a deletion in the El, E2b and/or 100K regions of the adenovirus genome. The adenovirus vectors can comprise vectors having the El, E2b and/or protease functions deleted. In some cases, such vectors have no other deletions. The adenovirus vectors can have the El and/or the E2b regions deleted, while the fiber genes have been modified by mutation or other alterations (for example to alter Ad tropism). Removal of genes from the E3 or E4 regions may be added to any of the adenovirus vectors mentioned. In certain embodiments, the adenovirus vector may be a gutted adenovirus vector.

[00210] Other regions of the Ad genome can be deleted. A "deletion" in a particular region of the Ad genome refers to a specific DNA sequence that is mutated or removed in such a way so as to prevent expression and/or function of at least one gene product encoded by that region (e.g., E2b functions of DNA polymerase or preterminal protein function). Deletions encompass deletions within exons encoding portions of proteins as well as deletions within promoter and leader sequences. A deletion within a particular region refers to a deletion of at least one base pair within that region of the Ad genome. More than one base pair can be deleted. For example, at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 base pairs can be deleted from a particular region. The deletion can be more than 150, 160, 170, 180, 190, 200, 250, or 300 base pairs within a particular region of the Ad genome. These deletions can prevent expression and/or function of the gene product encoded by the region. For example, a particular region of the Ad genome can include one or more point mutations such that one or more encoded proteins is non-functional. Such mutations include residues that are replaced with a different residue leading to a change in the amino acid sequence that result in a nonfunctional protein. Exemplary deletions or mutations in the Ad genome include one or more of Ela, Elb, E2a, E2b, E3, E4, LI, L2, L3, L4, L5, TP, POL, IV, and VA regions. Deleted adenovirus vectors can be made, for example, using recombinant techniques.

[00211] Ad vectors in certain embodiments can be successfully grown to high titers using an appropriate packaging cell line that constitutively expresses E2b gene products and products of any of the necessary genes that may have been deleted. HEK-293 -derived cells that not only

constitutively express the El and DNA polymerase proteins, but also the Ad -preterminal protein, can be used. E.C7 cells can be used, for example, to grow high titer stocks of the adenovirus vectors.

[00212] To delete critical genes from self-propagating adenovirus vectors, proteins encoded by the targeted genes can first be coexpressed in HEK-293 cells, or similar, along with El proteins. For example, those proteins which are non-toxic when coexpressed constitutively (or toxic proteins inducibly-expressed) can be selectively utilized. Coexpression in HEK-293 cells of the El and E4 genes is possible (for example utilizing inducible, not constitutive, promoters). The El and protein IX genes, a virion structural protein, can be coexpressed. Further coexpression of the El, E4, and protein IX genes is also possible. El and 100K genes can be expressed in trans-complementing cell lines, as can El and protease genes.

[00213] Cell lines coexpressing El and E2b gene products for use in growing high titers of E2b deleted Ad particles can be used. Useful cell lines constitutively express the approximately 140 kDa Ad-DNA polymerase and/or the approximately 90 kDa preterminal protein. Cell lines that have high-level, constitutive coexpression of the El, DNA polymerase, and preterminal proteins, without toxicity (e.g. E.C7), are desirable for use in propagating Ad for use in multiple vaccinations. These cell lines permit the propagation of adenovirus vectors deleted for the El, DNA polymerase, and preterminal proteins.

[00214] The recombinant Ad can be propagated using, for example, tissue culture plates containing E.C7 cells infected with Ad vector virus stocks at an appropriate MOI (e.g., 5) and incubated at 37 °C for 40-96 h. The infected cells can be harvested, resuspended in 10 mM Tris-Cl (pH 8.0), and sonicated, and the virus can be purified by two rounds of cesium chloride density centrifugation. The virus containing band can be desalted over a column, sucrose or glycerol can be added, and aliquots can be stored at -80 °C. Virus can be placed in a solution designed to enhance its stability, such as A195. The titer of the stock can be measured (e.g., by measurement of the optical density at 260 nm of an aliquot of the virus after lysis). Plasmid DNA, either linear or circular, encompassing the entire recombinant E2b deleted adenovirus vector can be transfected into E.C7, or similar cells, and incubated at 37 °C until evidence of viral production is present (e.g. cytopathic effect). Conditioned media from cells can be used to infect more cells to expand the amount of virus produced before purification. Purification can be accomplished, for example, by two rounds of cesium chloride density centrifugation or selective filtration. Virus may be purified by chromatography using commercially available products or custom chromatographic columns.

[00215] The compositions as described herein can comprise enough virus to ensure that cells to be infected are confronted with a certain number of viruses. Thus, some embodiments provide a stock of recombinant Ad, such as an RCA-free stock of recombinant Ad. Viral stocks can vary

considerably in titer, depending largely on viral genotype and the protocol and cell lines used to prepare them. Viral stocks can have a titer of at least about 10 6 , 10 7 , or 10 s pfu/mL, or higher, such as at least about 10 9 , 10 10 , 10 11 , or 10 12 pfu/mL. Depending on the nature of the recombinant virus and the packaging cell line, a viral stock can have a titer of even about 10 13 particles/ml or higher.

[00216] A replication defective adenovirus vector (e.g., SEQ. ID. NO.:3) can comprise a sequence encoding a target antigen, a fragment thereof, or a variant thereof, at a suitable position. In some embodiments, a replication defective adenovirus vector (e.g., SEQ. ID. NO.:3) can comprise a sequence encoding a target antigen described herein, or a fragment, a variant, or a variant fragment thereof, at a position replacing the nucleic acid sequence encoding a CEA or a variant CEA (e.g., SEQ. ID. NO.: l). In some embodiments, a replication defective adenovirus vector (e.g., SEQ. ID. NO.: 3) can comprise a sequence encoding a target antigen described herein, or a fragment, a variant, or a variant fragment thereof, at a position replacing the nucleic acid sequence encoding a CEA or a variant CEA (e.g., SEQ. ID. NO.:2). In some embodiments, a replication defective adenovirus vector (e.g., SEQ. ID. NO.:3) can comprise a sequence encoding a target antigen described herein, or a fragment, a variant, or a variant fragment thereof, at a position replacing the nucleic acid sequence encoding a MUCl or a variant MUCl (e.g., SEQ. ID. N0. 5, SEQ. ID. NO.:6 or SEQ. ID. NO.:9). In some embodiments, a replication defective adenovirus vector (e.g., SEQ. ID. NO.:3) can comprise a sequence encoding a target antigen described herein, or a fragment, a variant, or a variant fragment thereof, at a position replacing the nucleic acid sequence encoding a T or a variant T (e.g., SEQ. ID. NO : 7 or SEQ. ID. NO : 8).

Polynucleotides and Variants Encoding Antigen Targets

[00217] Certain embodiments provide nucleic acid sequences, also referred to herein as polynucleotides that encode one or more target antigens of interest, or fragments or variants thereof. As such, some embodiments provide polynucleotides that encode target antigens from any source as described further herein, vectors comprising such polynucleotides and host cells transformed or transfected with such expression vectors. In order to express a desired target antigen polypeptide, nucleotide sequences encoding the polypeptide, or functional equivalents, can be inserted into an appropriate Ad vector (e.g., using recombinant techniques). The appropriate adenovirus vector may contain the necessary elements for the transcription and translation of the inserted coding sequence and any desired linkers. Methods which are well known to those skilled in the art may be used to construct these adenovirus vectors containing sequences encoding a polypeptide of interest and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.

[00218] Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a target antigen polypeptide/protein/epitope or a portion thereof) or may comprise a sequence that encodes a variant, fragment, or derivative of such a sequence. Polynucleotide sequences can encode target antigen proteins. In some embodiments, polynucleotides represent a novel gene sequence optimized for expression in specific cell types that may substantially vary from the native nucleotide sequence or variant but encode a similar protein antigen.

[00219] In other related embodiments, polynucleotide variants have substantial identity to native sequences encoding proteins (e.g., target antigens of interest), for example those comprising at least 70% sequence identity, preferably at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a native polynucleotide sequence encoding the polypeptides (e.g., BLAST analysis using standard parameters). These values can be appropriately adjusted to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning and the like.

Polynucleotides can encode a protein comprising for example at least 70% sequence identity, preferably at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a protein sequence encoded by a native polynucleotide sequence.

[00220] Polynucleotides can comprise at least about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 11, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 or more contiguous nucleotides encoding a polypeptide (e.g., target protein antigens), and all intermediate lengths there between. "Intermediate lengths", in this context, refers to any length between the quoted values, such as 16, 17, 18, 19, etc.; 21, 22, 23, etc.; 30, 31, 32, etc.; 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through 200-500; 500-1,000, and the like. A polynucleotide sequence may be extended at one or both ends by additional nucleotides not found in the native sequence encoding a polypeptide, such as an epitope or heterologous target protein. This additional sequence may consist of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides or more, at either end of the disclosed sequence or at both ends of the disclosed sequence.

[00221] The polynucleotides, regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, expression control sequences,

polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol. Illustrative polynucleotide segments with total lengths of about 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10,000, about 500, about 200, about 100, about 50 base pairs in length, and the like, (including all intermediate lengths) are contemplated to be useful in many embodiments.

[00222] A mutagenesis approach, such as site-specific mutagenesis, can be employed to prepare target antigen sequences. Specific modifications in a polypeptide sequence can be made through mutagenesis of the underlying polynucleotides that encode them. Site-specific mutagenesis can be used to make mutants through the use of oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. For example, a primer comprising about 14 to about 25 nucleotides or so in length can be employed, with about 5 to about 10 residues on both sides of the junction of the sequence being altered. Mutations may be made in a selected polynucleotide sequence to improve, alter, decrease, modify, or otherwise change the properties of the

polynucleotide, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide.

[00223] Mutagenesis of polynucleotide sequences can be used to alter one or more properties of the encoded polypeptide, such as the immunogenicity of an epitope comprised in a polypeptide or the oncogenicity of a target antigen. Assays to test the immunogenicity of a polypeptide include, but are not limited to, T-cell cytotoxicity assays (CTL/chromium release assays), T-cell proliferation assays, intracellular cytokine staining, ELISA, ELISpot, etc. Other ways to obtain sequence variants of peptides and the DNA sequences encoding them can be employed. For example, recombinant vectors encoding the desired peptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.

[00224] Polynucleotide segments or fragments encoding the polypeptides as described herein may be readily prepared by, for example, directly synthesizing the fragment by chemical means.

Fragments may be obtained by application of nucleic acid reproduction technology, such as PCR, by introducing selected sequences into recombinant vectors for recombinant production.

[00225] A variety of vector/host systems may be utilized to contain and produce polynucleotide sequences. Exemplary systems include microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA vectors; yeast transformed with yeast vectors; insect cell systems infected with virus vectors (e.g., baculovirus); plant cell systems transformed with virus vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.

[00226] Control elements or regulatory sequences present in an Ad vector may include those non- translated regions of the vector-enhancers, promoters, and 5' and 3' untranslated regions. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, sequences encoding a polypeptide of interest may be ligated into an Ad transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing the polypeptide in infected host cells. In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.

[00227] Specific initiation signals may also be used to achieve more efficient translation of sequences encoding a polypeptide of interest (e.g., ATG initiation codon and adjacent sequences). Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used. Specific termination sequences, either for transcription or translation, may also be incorporated in order to achieve efficient translation of the sequence encoding the polypeptide of choice.

[00228] A variety of protocols for detecting and measuring the expression of polynucleotide- encoded products (e.g., target antigens), can be used (e.g., using polyclonal or monoclonal antibodies specific for the product). Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS). A two-site, monoclonal- based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on a given polypeptide may be preferred for some applications, but a competitive binding assay may also be employed.

[00229] The Ad vectors can comprise a product that can be detected or selected for, such as a reporter gene whose product can be detected, such as by fluorescence, enzyme activity on a chromogenic or fluorescent substrate, and the like, or selected for by growth conditions. Exemplary reporter genes include green fluorescent protein (GFP), β-galactosidase, chloramphenicol acetyltransferase (CAT), luciferase, neomycin phosphotransferase, secreted alkaline phosphatase (SEAP), and human growth hormone (HGH). Exemplary selectable markers include drug resistances, such as neomycin (G418), hygromycin, and the like.

[00230] The Ad vectors can also comprise a promoter or expression control sequence. The choice of the promoter will depend in part upon the targeted cell type and the degree or type of control desired. Promoters that are suitable include, without limitation, constitutive, inducible, tissue specific, cell type specific, temporal specific, or event-specific. Examples of constitutive or nonspecific promoters include the SV40 early promoter, the SV40 late promoter, CMV early gene promoter, bovine papilloma virus promoter, and adenovirus promoter. In addition to viral promoters, cellular promoters are also amenable and useful in some embodiments. In particular, cellular promoters for the so-called housekeeping genes are useful (e.g., β-actin). Viral promoters are generally stronger promoters than cellular promoters. Inducible promoters may also be used. These promoters include MMTV LTR, inducible by dexamethasone, metallothionein, inducible by heavy metals, and promoters with cAMP response elements, inducible by cAMP, heat shock promoter. By using an inducible promoter, the nucleic acid may be delivered to a cell and will remain quiescent until the addition of the inducer. This allows further control on the timing of production of the protein of interest. Event-type specific promoters (e.g., HIV LTR) can be used, which are active or upregulated only upon the occurrence of an event, such as tumorigenicity or viral infection, for example. The HIV LTR promoter is inactive unless the tat gene product is present, which occurs upon viral infection. Some event-type promoters are also tissue-specific. Preferred event-type specific promoters include promoters activated upon viral infection.

[00231] Examples of promoters include promoters for a-fetoprotein, a-actin, myo D,

carcinoembryonic antigen, VEGF-receptor; FGF receptor; TEK or tie 2; tie; urokinase receptor; E- and P-selectins; VCAM-1; endoglin; endosialin; αν-β3 integrin; endothelin-1; ICAM-3; E9 antigen; von Willebrand factor; CD44; CD40; vascular-endothelial cadherin; notch 4, high molecular weight melanoma-associated antigen; prostate specific antigen- 1, probasin, FGF receptor, VEGF receptor, erb B2; erb B3; erb B4; MUC-1; HSP-27; int-1; int-2, CEA, HBEGF receptor; EGF receptor;

tyrosinase, MAGE, IL-2 receptor; prostatic acid phosphatase, probasin, prostate specific membrane antigen, a-crystallin, PDGF receptor, integrin receptor, a-actin, SMI and SM2 myosin heavy chains, calponin-hl, SM22 a-angiotensin receptor, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, immunoglobulin heavy chain, immunoglobulin light chain, and CD4.

[00232] Repressor sequences, negative regulators, or tissue-specific silencers may be inserted to reduce non-specific expression of the polynucleotide. Multiple repressor elements may be inserted in the promoter region. Repression of transcription is independent of the orientation of repressor elements or distance from the promoter. One type of repressor sequence is an insulator sequence. Such sequences inhibit transcription and can silence background transcription. Negative regulatory elements can be located in the promoter regions of a number of different genes. The repressor element can function as a repressor of transcription in the absence of factors, such as steroids, as does the NSE in the promoter region of the ovalbumin gene. These negative regulatory elements can bind specific protein complexes from oviduct, none of which are sensitive to steroids. Three different elements are located in the promoter of the ovalbumin gene. Oligonucleotides

corresponding to portions of these elements can repress viral transcription of the TK reporter. One of the silencer elements shares sequence identity with silencers in other genes (TCTCTCCNA (SEQ ID NO: 11)).

[00233] Elements that increase the expression of the desired target antigen can be incorporated into the nucleic acid sequence of the Ad vectors described herein. Exemplary elements include internal ribosome binding sites (IRESs). IRESs can increase translation efficiency. As well, other sequences may enhance expression. For some genes, sequences especially at the 5' end may inhibit transcription and/or translation. These sequences are usually palindromes that can form hairpin structures. In some cases, such sequences in the nucleic acid to be delivered are deleted. Expression levels of the transcript or translated product can be assayed to confirm or ascertain which sequences affect expression. Transcript levels may be assayed by any known method, including Northern blot hybridization, RNase probe protection and the like. Protein levels may be assayed by any known method, including ELISA.

Antigen-Specific Immunotherapies and Vaccines

[00234] Certain embodiments provide single antigen or combination antigen immunization against MUC1, MUClc, MUCln, T, or CEA utilizing such vectors and other vectors as provided herein Certain embodiments provide therapeutic vaccines against MUC1, MUClc, MUCln, T, or CEA. Certain embodiments provide prophylactic vaccines against MUCl, MUClc, MUCln, T, or CEA. Further, in various embodiments, the composition and methods provide herein can lead to clinical responses, such as altered disease progression or life expectancy.

[00235] Ad5 [E1-] vectors encoding a variety of antigens can be used to efficiently transduce 95% of ex vivo exposed DCs to high titers of the vector. In certain embodiments, increasing levels of foreign gene expression in the DC was found to correlate with increasing multiplicities of infection (MOI) with the vector. DCs infected with Ad5 [E1-] vectors can encode a variety of antigens (including the tumor antigens MART-1, MAGE-A4, DF3/MUC1, p53, hugplOO melanoma antigen, polyoma virus middle -T antigen) that have the propensity to induce antigen specific CTL responses, have an enhanced antigen presentation capacity, and/or have an improved ability to initiate T-cell proliferation in mixed lymphocyte reactions. Immunization of animals with dendritic cells (DCs) previously transduced by Ad5 vectors encoding tumor specific antigens can be used to induce significant levels of protection for the animals when challenged with tumor cells expressing the respective antigen. Interestingly, intra-tumoral injection of Ads encoding IL-7 is less effective than injection of DCs transduced with IL-7 encoding Ad5 vectors at inducing antitumor immunity. Ex vivo transduction of DCs by Ad5 vectors is contemplated in certain embodiments. Ex vivo DC transduction strategies can been used to induce recipient host tolerance. For example, Ad5 mediated delivery of the CTLA4Ig into DCs can block interactions of the DCs CD80 with CD28 molecules present on T-cells.

[00236] Ad5 vector capsid interactions with DCs may trigger several beneficial responses, which may be enhancing the propensity of DCs to present antigens encoded by Ad5 vectors. For example, immature DCs, though specialized in antigen uptake, are relatively inefficient effectors of T-cell activation. DC maturation coincides with the enhanced ability of DCs to drive T-cell immunity. In some instances, the compositions and methods take advantage of an Ad5 infection resulting in direct induction of DC maturation Ad vector infection of immature bone marrow derived DCs from mice may upregulate cell surface markers normally associated with DC maturation (MHC I and II, CD40, CD80, CD86, and ICAM-1) as well as down-regulation of CD1 lc, an integrin down regulated upon myeloid DC maturation. In some instances, Ad vector infection triggers IL-12 production by DCs, a marker of DC maturation. Without being bound by theory, these events may possibly be due to Ad5 triggered activation of NF-κΒ pathways. Mature DCs can be efficiently transduced by Ad vectors, and do not lose their functional potential to stimulate the proliferation of naive T-cells at lower MOI, as demonstrated by mature CD83+ human DC (derived from peripheral blood monocytes). However, mature DCs may also be less infectable than immature ones. Modification of capsid proteins can be used as a strategy to optimize infection of DC by Ad vectors, as well as enhancing functional maturation, for example using the CD40L receptor as a viral vector receptor, rather than using the normal CAR receptor infection mechanisms.

[00237] In some embodiments the compositions and methods comprising an Ad5 [E1-, E2b-] vector(s) MUC1, T and CEA vaccine have effects of increased overall survival (OS) within the bounds of technical safety. In some embodiments the compositions and methods comprising an Ad5 [E1-, E2b-] vector(s) MUClc, T and CEA vaccine have effects of increased overall survival (OS) within the bounds of technical safety. In certain embodiments the compositions and methods comprising an Ad5 [E1-, E2b-] vector(s) MUCln, T and CEA vaccine have effects of increased overall survival (OS) within the bounds of technical safety.

[00238] In some embodiments, the antigen targets are associated with benign tumors. In some embodiments, the antigens targeted are associated with pre-cancerous tumors.

[00239] In some embodiments, the antigens targeted are associated with carcinomas, in situ carcinomas, metastatic tumors, neuroblastoma, sarcomas, myosarcoma, leiomyosarcoma, retinoblastoma, hepatoma, rhabdomyosarcoma, plasmocytomas, adenomas, gliomas, thymomas, or osteosarcoma. In some embodiments, the antigens targeted are associated with a specific type of cancer such as neurologic cancers, brain cancer, thyroid cancer, head and neck cancer, melanoma, leukemia, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL), non-Hodgkin's lymphoma, multiple myeloma, Hodgkin's disease, breast cancer, bladder cancer, prostate cancer, colorectal cancer, colon cancer, kidney cancer, renal cell carcinoma, pancreatic cancer, esophageal cancer, lung cancer, mesothelioma, ovarian cancer, cervical cancer, endometrial cancer, uterine cancer, germ cell tumors, testicular cancer, gastric cancer, or other cancers, or any clinical (e.g. TNM, Histopathological, Staging or Grading systems or a combination thereof) or molecular subtype thereof. In some embodiments, the antigens targeted are associated with a specific clinical or molecular subtype of cancer. By way of example, breast cancer can be divided into at least four molecular subtypes including Luminal A, Luminal B, Triple negative/basal -like, and HER2 type. By way of example, Prostate cancer can be subdivided TNM, Gleason score, or molecular expression of the PSA protein.

[00240] As noted above, the adenovirus vectors comprise nucleic acid sequences that encode one or more target proteins or antigens of interest. In this regard, the vectors may contain nucleic acid encoding 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more different target antigens of interest. The target antigens may be a full length protein or may be a fragment (e.g., an epitope) thereof. The adenovirus vectors may contain nucleic acid sequences encoding multiple fragments or epitopes from one target protein of interest or may contain one or more fragments or epitopes from numerous different target proteins of interest. A target antigen may comprise any substance against which it is desirable to generate an immune response but generally, the target antigen is a protein. A target antigen may comprise a full length protein, a subunit of a protein, an isoform of a protein, or a fragment thereof that induces an immune response (i.e., an immunogenic fragment). A target antigen or fragment thereof may be modified, e.g., to reduce one or more biological activities of the target antigen or to enhance its immunogenicity. The target antigen or target protein can be MUC1, MUClc, MUCln, T, CEA, or any combination thereof.

[00241] In certain embodiments, immunogenic fragments bind to an MHC class I or class II molecule. An immunogenic fragment may "bind to" an MHC class I or class II molecule if such binding is detectable using any assay known in the art. For example, the ability of a polypeptide to bind to MHC class I may be evaluated indirectly by monitoring the ability to promote incorporation of 125 I labeled β-2-microglobulin (β-2ιη) into MHC class 1/ 2m/peptide heterotrimeric complexes. Alternatively, functional peptide competition assays that are known in the art may be employed. Immunogenic fragments of polypeptides may generally be identified using well known techniques. Representative techniques for identifying immunogenic fragments include screening polypeptides for the ability to react with antigen-specific antisera and/or T-cell lines or clones. An immunogenic fragment of a particular target polypeptide is a fragment that reacts with such antisera and/or T-cells at a level that is not substantially less than the reactivity of the full length target polypeptide (e.g., in an ELISA and/or T-cell reactivity assay). In other words, an immunogenic fragment may react within such assays at a level that is similar to or greater than the reactivity of the full length polypeptide. Such screens may be performed using methods known in the art.

[00242] In some embodiments, the viral vectors comprise heterologous nucleic acid sequences that encode one or more proteins, variants thereof, fusions thereof, or fragments thereof, that can modulate the immune response. In some embodiments, the viral vector encodes one or more antibodies against specific antigens, such as anthrax protective antigen, permitting passive immunotherapy. In some embodiments, the viral vectors comprise heterologous nucleic acid sequences encoding one or more proteins having therapeutic effect (e.g., anti-viral, anti -bacterial, anti-parasitic, or anti-tumor function). In some embodiments the Second Generation E2b deleted adenovirus vectors comprise a heterologous nucleic acid sequence. In some embodiments, the heterologous nucleic acid sequence is CEA, MUCl, MUClc, MUCln, T, a variant, a portion, or any combination thereof.

[00243] Target antigens include, but are not limited to, antigens derived from a variety of tumor proteins. In some embodiments, parts or variants of tumor proteins are employed as target antigens. In some embodiments, parts or variants of tumor proteins being employed as target antigens have a modified, for example, increased ability to effect and immune response against the tumor protein or cells containing the same. A vaccine can vaccinate against an antigen. A vaccine can also target an epitope. An antigen can be a tumor cell antigen. An epitope can be a tumor cell epitope. Such a tumor cell epitope may be derived from a wide variety of tumor antigens, such as antigens from tumors resulting from mutations, shared tumor specific antigens, differentiation antigens, and antigens overexpressed in tumors. Tumor-associated antigens (TAAs) may be antigens not normally expressed by the host; they can be mutated, truncated, misfolded, or otherwise abnormal

manifestations of molecules normally expressed by the host; they can be identical to molecules normally expressed but expressed at abnormally high levels; or they can be expressed in a context or environment that is abnormal. Tumor-associated antigens may be, for example, proteins or protein fragments, complex carbohydrates, gangliosides, haptens, nucleic acids, other biological molecules or any combinations thereof.

[00244] Illustrative useful tumor proteins include, but are not limited to any one or more of, CEA, human epidermal growth factor receptor 2 (HER2/neu), human epidermal growth factor receptor 3 (HER3), Human papillomavirus (HPV) E6, HPV E7, MUCl, Prostate-specific antigen (PSA), PSMA, WT1, p53, MAGE-Al, MAGE-A2, MAGE- A3, MAGE-A4, MAGE-A6, MAGE- A 10, MAGE-A12, BAGE, DAM-6, DAM-10, GAGE-1, GAGE-2, GAGE- 8, GAGE-3, GAGE-4, GAGE- 5, GAGE-6, GAGE-7B, NA88-A, NY-ESO-1, MART-1, MC1R, GplOO, PSA, PSM, Tyrosinase, TRP-1, TRP-2, ART-4, CAMEL, CEA, Cyp-B, Her2/neu, BRCA1, Brachyury, Brachyury (TIVS7- 2, polymorphism), Brachyury (rVS7 T/C polymorphism), T Brachyury, T, hTERT, hTRT, iCE, MUCl, MUCl (VNTR polymorphism), MUClc, MUCln, MUC2, PRAME, P15, RU1, RU2, SART-1, SART-3, AFP, β-catenin/m, Caspase-8/m, CDK-4/m, ELF2M, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, Myosin/m, RAGE, SART-2, TRP-2/INT2, 707-AP, Annexin Π, CDC27/m, TPI/mbcr-abl, ETV6/AML, LDLR/FUT, Pml/RARa, and TEL/AML1.

[00245] In some embodiments, the viral vector comprises a target antigen sequence encoding a modified polypeptide selected from CEA, human epidermal growth factor receptor 2 (HER2/neu), human epidermal growth factor receptor 3 (HER3), Human papillomavirus (HPV) E6, HPV E7, MUCl, Prostate-specific antigen (PSA), PSMA (i.e., PSM), WT1, p53, MAGE-Al, MAGE-A2, MAGE- A3, MAGE-A4, MAGE-A6, MAGE-Al 0, MAGE-Al 2, BAGE, DAM-6, DAM-10, GAGE- 1, GAGE-2, GAGE- 8, GAGE-3, GAGE-4, GAGE- 5, GAGE-6, GAGE-7B, NA88-A, NY-ESO-1, MART-1, MC1R, GplOO, Tyrosinase, TRP-1, TRP-2, ART-4, CAMEL, Cyp-B, BRCA1,

Brachyury, Brachyury (TIVS7-2, polymorphism), Brachyury (IVS7 T/C polymorphism), T

Brachyury, T, hTERT, hTRT, iCE, MUC1 (VNTR polymorphism), MUClc, MUCln, MUC2, PRAME, P15, RU1, RU2, SART-1, SART-3, AFP, β-catenin/m, Caspase-8/m, CDK-4/m, ELF2M, GnT-V, G250, HSP70-2M, HST-2, KIAA0205, MUM-1, MUM-2, MUM-3, Myosin/m, RAGE, SART-2, TRP-2/INT2, 707- AP, Annexin Π, CDC27/m, TPI/mbcr-abl, ETV6/AML, LDLR/FUT, Pml/RARa, and TEL/AML1, wherein the polypeptide or a fragment thereof has at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to the corresponding native sequence.

[00246] Additional illustrative useful tumor proteins useful include, but are not limited to any one or more of alpha-actinin-4, ARTC1, CAR-ABL fusion protein (b3a2), B-RAF, CASP-5, CASP-8, beta-catenin, Cdc27, CDK4, CDKN2A, COA-1, dek-can fusion protein, EFTUD2, Elongation factor 2, ETV6-AML1 fusion protein, FLT3-ITD, FNl, GPNMB, LDLR-fucosyltransferase fusion protein, HLA-A2d, HLA-A1 Id, hsp70-2, KIAAO205, MART2, ME1, MUM-lf, MUM-2, MUM-3, neo- PAP, Myosin class I, NFYC, OGT, OS-9, p53, pml-RARalpha fusion protein, PRDX5, PTPRK, K- ras, N-ras, RBAF600, SIRT2, SNRPD1, SYT-SSX1- or -SSX2 fusion protein, TGF-betaRII, tnosephosphate isomerase, BAGE-1, GnTVf, HERV-K-MEL, KK-LC-1, KM-HN-1, LAGE-1, MAGE-A9, MAGE-C2, mucink, NA-88, NY-ESO-l/LAGE-2, SAGE, Spl7, SSX-2, SSX-4, TAG- 1, TAG-2, TRAG-3, TRP2-INT2g, XAGE-lb, gpl00/Pmell7, Kal krein 4, mammaglobin-A, Melan- A/MART- 1 , NY-BR-1, OA1, PSA, RAB38/NY-MEL-1, TRP-l/gp75, TRP-2, tyrosinase, adipophilm, AFM-2, ALDH1A1, BCLX (L), BCMA, BING-4, CPSF, cyclin Dl, DKK1, ENAH (hMena), EP-CAM, EphA3, EZH2, FGF5, G250/MN/CAIX, IL13Ralpha2, intestinal carboxyl esterase, alpha fetoprotein, M-CSFT, MCSP, mdm-2, MMP-2, PBF, PRAME, RAGE-1, RGS5, RNF43, RU2AS, secernin 1, SOX10, STEAP1, survivin, Telomerase, and/or VEGF.

[00247] Tumor-associated antigens may be antigens from infectious agents associated with human malignancies. Examples of infectious agents associated with human malignancies include Epstein- Barr virus, Helicobacter pylori, Hepatitis B virus, Hepatitis C virus, Human heresvirus-8, Human immunodeficiency virus, Human papillomavirus, Human T-cell leukemia virus, liver flukes, and Schistosoma haematobium.

CEA Antigen Targets

[00248] CEA represents an attractive target antigen for immunotherapy since it is over-expressed in nearly all colorectal cancers and pancreatic cancers, and is also expressed by some lung and breast cancers, and uncommon tumors such as medullary thyroid cancer, but is not expressed in other cells of the body except for low-level expression in gastrointestinal epithelium. CEA contains epitopes that may be recognized in an MHC restricted fashion by T-cells. [00249] It was discovered that multiple homologous immunizations with Ad5 [E1-, E2b-]- CEA(6D), encoding the tumor antigen CEA, induced CEA-specific cell-mediated immune (CMI) responses with antitumor activity in mice despite the presence of pre-existing or induced Ad5- neutralizing antibody. In the present phase I/II study, cohorts of patients with advanced colorectal cancer were immunized with escalating doses of Ad5 [E1-, E2b-]-CEA(6D). CEA-specific CMI responses were observed despite the presence of pre-existing Ad5 immunity in a majority (61.3%) of patients. Importantly, there was minimal toxicity, and overall patient survival (48% at 12 months) was similar regardless of pre-existing Ad5 neutralizing antibody titers. The results demonstrate that, in cancer patients, the novel Ad5 [E1-, E2b-] gene delivery platform generates significant CMI responses to the tumor antigen CEA in the setting of both naturally acquired and immunization- induced Ad5specific immunity.

[00250] CEA antigen specific CMI can be, for example, greater than 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 5000, 10000, or more IFN-γ spot forming cells (SFC) per 10 6 peripheral blood mononuclear cells (PBMC). In some embodiments, the immune response is raised in a human subject with a preexisting inverse Ad5 neutralizing antibody titer of greater than 50, 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 6000, 7000, 8000, 9000, 1000, 12000, 15000 or higher. The immune response may comprise a cell-mediated immunity and/or a humoral immunity as described herein. The immune response may be measured by one or more of intracellular cytokine staining (ICS), ELISpot, proliferation assays, cytotoxic T-cell assays including chromium release or equivalent assays, and gene expression analysis using any number of polymerase chain reaction (PCR) or RT-PCR based assays, as described herein and to the extent they are available to a person skilled in the art, as well as any other suitable assays known in the art for measuring immune response.

[00251] In some embodiments, the replication defective adenovirus vector comprises a modified sequence encoding a subumt with at least 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to a wild-type subunit of the polypeptide.

[00252] The immunogenic polypeptide may be a mutant CEA or a fragment thereof. In some embodiments, the immunogenic polypeptide comprises a mutant CEA with an Asn->Asp substitution at position 610. In some embodiments, the replication defective adenovirus vector comprises a sequence encoding a polypeptide with at least 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to the immunogenic polypeptide. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the sequence of SEQ ID NO: 1.

[00253] In some embodiments, the sequence encoding the immunogenic polypeptide comprises a sequence with at least 70% 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to SEQ. ID. NO: 1 or a sequence generated from SEQ ID NO: 1 by alternative codon replacements. In some embodiments, the immunogenic polypeptide encoded by the adenovirus vectors comprise up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, or more point mutations, such as single amino acid substitutions or deletions, as compared to a wild-type human CEA sequence.

[00254] In some embodiments, the immunogenic polypeptide comprises a sequence from SEQ ID NO.:2 or a modified version, e.g., comprising up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, or more point mutations, such as single amino acid substitutions or deletions, of SEQ ID NO.: 2.

[00255] Members of the CEA gene family are subdivided into three subgroups based on sequence similarity, developmental expression patterns and their biological functions: the CEA-related Cell Adhesion Molecule (CEACAM) subgroup containing twelve genes (CEACAM1, CEACAM3- CEACAM8, CEACAM 16 and CEACAM 18-CEACAM21), the Pregnancy Specific Glycoprotein (PSG) subgroup containing eleven closely related genes (PSG1-PSG11) and a subgroup of eleven pseudogenes (CEACAMP1-CEACAMP11). Most members of the CEACAM subgroup have similar structures consist of an extracellular Ig-like domains composed of a single N-terminal V-set domain, with structural homology to the immunoglobulin variable domains, followed by varying numbers of C2-set domains of A or B subtypes, a transmembrane domain and a cytoplasmic domain. There are two members of CEACAM subgroup (CEACAM 16 and CEACAM20) that show a few exceptions in the organization of their structures. CEACAM 16 contains two Ig-like V-type domains at its N and C termini and CEACAM20 contains a truncated Ig-like V-type 1 domain. The CEACAM molecules can be anchored to the cell surface via their transmembrane domains (CEACAM5 thought

CEACAM8) or directly linked to glycophosphatidylinositol (GPI) lipid moiety (CEACAM5,

CEACAM18 thought CEACAM21).

[00256] CEA family members are expressed in different cell types and have a wide range of biological functions. CEACAMs are found prominently on most epithelial cells and are present on different leucocytes. In humans, CEACAMl, the ancestor member of CEA family, is expressed on the apical side of epithelial and endothelial cells as well as on lymphoid and myeloid cells.

CEACAMl mediates cell-cell adhesion through hemophilic (CEACAMl to CEACAMl) as well as heterothallic (e.g., CEACAMl to CEACAMS) interactions. In addition, CEACAMl is involved in many other biological processes, such as angiogenesis, cell migration, and immune functions.

CEACAMS and CEACAM4 expression is largely restricted to granulocytes, and they are able to convey uptake and destruction of several bacterial pathogens including Neisseria, Moraxella, and Haemophilus species.

[00257] Thus, in various embodiments, compositions and methods relate to raising an immune response against a CEA, selected from the group consisting of CEACAMl, CEACAM3,

CEACAM4, CEACAM5, CEACAM6, CEACAM7, CEACAM8, CEACAMl 6, CEACAMl 8, CEACAMl 9, CEACAM20, CEACAM21, PSG1, PSG2, PSG3, PSG4, PSG5, PSG6, PSG7, PSG8, PSG9, and PSGl 1. An immune response may be raised against cells, e.g. cancer cells, expressing or overexpressing one or more of the CEAs, using the methods and compositions. In some

embodiments, the overexpression of the one or more CEAs in such cancer cells is over 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 fold or more compared to non-cancer cells.

[00258] In certain embodiments, the CEA antigen used herein is a wild-type CEA antigen or a modified CEA antigen having a least a mutation in YLSGA LNL (SEQ ID NO:4), a CAP1 epitope of CEA. The mutation can be conservative or non-conservative, substitution, addition, or deletion. In certain embodiments, the CEA antigen used herein has an amino acid sequence set forth in YLSGADL L (SEQ ID NO: 10), a mutated CA 1 epitope. In further embodiments, the first replication-defective vector or a replication-defective vector that express CEA has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%), 100%) identical to any portion of SEQ ID NO: 3 (the predicted sequence of an adenovirus vector express a modified CEA antigen), such as positions 1057 to 3165 of SEQ ID NO:3 or full-length SEQ ID NO:3.

Mucin Family Antigen Targets

[00259] The human mucin family (MUCl to MUC21) includes secreted and transmembrane mucins that play a role in forming protective mucous barriers on epithelial surfaces in the body. These proteins function in to protecting the epithelia lining the respiratory, gastrointestinal tracts, and lining ducts in important organs such as, for example the mammary gland, liver, stomach, pancreas, and kidneys.

[00260] MUCl (CD227) is a TAA that is over-expressed on a majority of human carcinomas and several hematologic malignancies. MUCl (GenBank: X80761.1, NCBI: NM_001204285.1) and activates many important cellular pathways known to be involved in human disease. MUCl is a heterodimeric protein formed by two subunits that is commonly overexpressed in several human cancers. MUCl undergoes autoproteolysis to generate two subunits MUCln and MUClc that, in turn, form a stable noncovalent heterodimer.

[00261] The MUCl C-terminal subunit (MUClc) can comprise a 58 aa extracellular domain (ED), a 28 aa transmembrane domain (TM) and a 72 aa cytoplasmic domain (CD). The MUClc also can contains a "CQC" motif that can allow for dimerization of MUCl and it can also impart oncogenic function to a cell. In some cases MUCl can in part oncogenic function through inducing cellular signaling via MUClc. MUClc can interact with EGFR, ErbB2 and other receptor tyrosine kinases and contributing to the activation of the PI3K→AKT and MEK→ERK cellular pathways. In the nucleus, MUClc activates the Wnt/ -catenin, STAT and NF-κΒ RelA cellular pathways. In some cases MUCl can impart oncogenic function through inducing cellular signaling via MUCln. The MUCl N-terminal subunit (MUCln) can comprise variable numbers of 20 amino acid tandem repeats that can be glycosylated. MUCl is normally expressed at the surface of glandular epithelial cells and is over-expressed and aberrantly glycosylated in carcinomas. MUC1 is a TAA that can be utilized as a target for tumor immunotherapy. Several clinical trials have been and are being performed to evaluate the use of MUC1 in immunotherapeutic vaccines. Importantly, these trials indicate that immunotherapy with MUC1 targeting is safe and may provide survival benefit.

[00262] However, clinical trials have also shown that MUC1 is a relatively poor immunogen. To overcome this, the inventors have identified a T lymphocyte immune enhancer peptide sequence in the C terminus region of the MUC1 oncoprotein (MUCl-C or MUClc). Compared with the native peptide sequence, the agonist in their modified MUCl-C (a) bound HLA-A2 at lower peptide concentrations, (b) demonstrated a higher avidity for HLA-A2, (c) when used with antigen- presenting cells, induced the production of more IFN-γ by T-cells than with the use of the native peptide, and (d) was capable of more efficiently generating MUCl -specific human T-cell lines from cancer patients. Importantly, T-cell lines generated using the agonist epitope were more efficient than those generated with the native epitope for the lysis of targets pulsed with the native epitope and in the lysis of HLA-A2 human tumor cells expressing MUCl. Additionally, the inventors have identified additional CD8+ cytotoxic T lymphocyte immune enhancer agonist sequence epitopes of MUCl-C.

[00263] Certain embodiments provide a potent MUCl-C modified for immune enhancer capability (mMUCl-C or MUCl-C or MUClc). Certain embodiments provide a potent MUCl-C modified for immune enhancer capability incorporated it into a recombinant Ad5 [E1-, E2b-] platform to produce a new and more potent immunotherapeutic vaccine. For example, the immunotherapeutic vaccine can be Ad5 [E1-, E2b-]-mMUCl-C for treating MUCl expressing cancers or infectious diseases.

[00264] Post-translational modifications play an important role in controlling protein function in the body and in human disease. For example, in addition to proteolytic cleavage discussed above, MUCl can have several post-translational modifications such as glycosylation, sialylation, palmitoylation, or a combination thereof at specific amino acid residues. Provided herein are immunotherapies targeting glycosylation, sialylation, phosphorylation, or palmitoylation

modifications of MUCl.

[00265] MUCl can be highly glycosylated (N- and O-linked carbohydrates and sialic acid at varying degrees on serine and threonine residues within each tandem repeat, ranging from mono- to penta-glycosylation). Differentially O-glycosylated in breast carcinomas with 3,4-linked GlcNAc. N- glycosylation consists of high-mannose, acidic complex-type and hybrid glycans in the secreted form MUCl/SEC, and neutral complex-type in the transmembrane form, MUC1/TM.4. Certain embodiments provide immunotherapies targeting differentially O-glycosylated forms of MUCl.

[00266] Further, MUCl can be sialylated. Membrane-shed glycoproteins from kidney and breast cancer cells have preferentially sialyated core 1 structures, while secreted forms from the same tissues display mainly core 2 structures. The O-glycosylated content is overlapping in both these tissues with terminal fucose and galactose, 2- and 3 -linked galactose, 3- and 3,6-linked GalNAc-ol and 4-linked GlcNAc predominating. Certain embodiments provide immunotherapies targeting various sialylation forms of MUCl. Dual palmitoylation on cysteine residues in the CQC motif is required for recycling from endosomes back to the plasma membrane. Certain embodiments provide for immunotherapies targeting various palmitoylation forms of MUCl.

[00267] Phosphorylation can affect MUCl 's ability to induces specific cell signaling responses that are important for human health. Certain embodiments provide for immunotherapies targeting various phosphorylated forms of MUCl. For example, MUCl can be phosphorylated on tyrosine and serine residues in the C-terminal domain. Phosphorylation on tyrosines in the C-terminal domain can increase nuclear location of MUCl and β-catenin. Phosphorylation by PKC delta can induce binding of MUCl to β-catenin/CTNNBl and decrease formation of β-catenin/E-cadherin complexes. Src- mediated phosphorylation of MUCl can inhibits interaction with GSK3B. Src- and EGFR-mediated phosphorylation of MUCl on Tyr-1229 can increase binding to β-catenin/CTNNBl. GSK3B- mediated phosphorylation of MUCl on Ser-1227 can decrease this interaction but restores the formation of the β-cadherin/E-cadherin complex. PDGFR-mediated phosphorylation of MUCl can increase nuclear colocalization of MUCl CT and CTNNB1. Certain embodiments provide immunotherapies targeting different phosphorylated forms of MUCl, MUClc and MUCln known to regulate its cell signaling abilities.

[00268] The disclosure provides for immunotherapies that modulate MUClc cytoplasmic domain and its functions in the cell. The disclosure provides for immunotherapies that comprise modulating a CQC motif in MUClc. The disclosure provides for immunotherapies that comprise modulating the extracellular domain (ED), the transmembrane domain (TM), the cytoplasmic domain (CD) of MUClc, or a combination thereof. The disclosure provides for immunotherapies that comprise modulating MUClc's ability to induce cellular signaling through EGFR, ErbB2 or other receptor tyrosine kinases. The disclosure provides for immunotherapies that comprise modulating MUClc's ability to induce PI3K→AKT, MEK→ERK, Wnt^-catemn, STAT, NF-κΒ RelA cellular pathways, or combination thereof. In some embodiments, the MUClc immunotherapy can further comprise CEA.

[00269] The disclosure also provides for immunotherapies that modulate MUCln and its cellular functions. The disclosure also provides for immunotherapies comprising tandem repeats of MUCln, the glycosylation sites on the tandem repeats of MUCln, or a combination thereof. In some embodiments, the MUCln immunotherapy further comprises CEA.

[00270] The disclosure also provides vaccines comprising MUCln, MUClc, CEA, or a combination thereof. The disclosure provides vaccines comprising MUClc and CEA. The disclosure also provides vaccines targeting MUCln and CEA. In some embodiments, the antigen combination is contained in one vector as provided herein. In some embodiments, the antigen combination is contained in a separate vector as provided herein.

[00271] Some embodiments relate to a replication defective adenovirus vector of serotype 5 comprising a sequence encoding an immunogenic polypeptide. The immunogenic polypeptide may be an isoform of MUCl or a subunit or a fragment thereof. In some embodiments, the replication defective adenovirus vector comprises a sequence encoding a polypeptide with at least 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to the immunogenic polypeptide. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the sequence of SEQ. ID. NO.: 5. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the following sequence identified by SEQ. ID. NO.: 6. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the following sequence identified by SEQ. ID. NO.:9. In some embodiments, the sequence encoding the immunogenic polypeptide comprises a sequence with at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to SEQ. ID. NO:5, SEQ. ID. NO:6, SEQ. ID. NO.:9 or a sequence generated from SEQ. ID. N0. 5, SEQ. ID. NO:6, SEQ. ID. NO.: 9 by alternative codon replacements. In some embodiments, the immunogenic polypeptide encoded by the adenovirus vectors described herein comprising up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, or more point mutations, such as single amino acid substitutions or deletions, as compared to a wild-type human MUCl sequence.

[00272] In certain embodiments, the MUCl antigen used herein is a wild-type MUCl antigen or a modified MUCl antigen. In certain embodiments, the modified MUCl antigen has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to SEQ ID NO:9 (a mutated MUCl protein sequence). In certain embodiments, the MUC-1 antigen is a modified antigen having one or more mutations at positions 93, 141-142, 149-151, 392, 404, 406, 422, 430-431, 444-445, or 460 of SEQ ID NO:9. The mutation can be conservative or non-conservative, substitution, addition, or deletion. In further embodiments, the MUC-1 antigen binds to HLA-A2, HLA-A3, HLA-A24, or a combination thereof. In certain embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO:5 (MUC_1 wild-type nucleotide sequence). In further embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to SEQ ID NO: 6 (a mutated MUCl nucleotide sequence). In certain embodiments, the third replication-defective vector or a replication-defective vector that express MUCl has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to any portion of or full-length SEQ ID NO: 14 (the predicted sequence of an adenovirus vector express a modified CEA antigen), such as positions 1033-2858 of SEQ ID NO: 14.

Brachyury Antigen Targets

[00273] Certain embodiment provide immunotherapies that comprise one or more antigens to Brachyury. Brachyury (also known as the "T" protein in humans) is a member of the T-box family of transcription factors that play key roles during early development, mostly in the formation and differentiation of normal mesoderm and is characterized by a highly conserved DNA-binding domain designated as T-domain. The epithelial to mesenchymal transition (EMT) is a key step during the progression of primary tumors into a metastatic state in which Brachyury plays a crucial role. The expression of Brachyury in human carcinoma cells induces changes characteristic of EMT, including up-regulation of mesenchymal markers, down-regulation of epithelial markers, and an increase in cell migration and invasion. Conversely, inhibition of Brachyury resulted in down- regulation of mesenchymal markers and loss of cell migration and invasion and diminished the ability of human tumor cells to form metastases. Brachyury can function to mediate epithelial - mesenchymal transition and promotes invasion.

[00274] The disclosure also provides for immunotherapies that modulate Brachyury effect on epithelial -mesenchymal transition function in cell proliferation diseases, such as cancer. The disclosure also provides for immunotherapies that modulate Brachyury' s ability to promote invasion in cell proliferation diseases, such as cancer. The disclosure also provides for immunotherapies that modulate the DNA binding function of T-box domain of Brachyury. In some embodiments, the Brachyury immunotherapy can further comprise one or more antigens to CEA or MUCl, MUClc or MUCln.

[00275] Brachyury expression is nearly undetectable in most normal human tissues and is highly restricted to human tumors and often overexpressed making it an attractive target antigen for immunotherapy. In human, Brachyury is encoded by the T gene (GenBank: AJ001699.1, NCBI: NM_003181.3). There are at least two different isoforms produced by alternative splicing found in humans. Each isoform has a number of natural variants.

[00276] Brachyury is immunogenic and Brachyury-specific CD8+ T-cells expanded in vitro can lyse Brachyury expressing tumor cells. These features of Brachyury make it an attractive TAA for immunotherapy. The Brachyury protein is a T-box transcription factor. It can bind to a specific DNA element, a near palindromic sequence "TCACACCT" through a region in its N-terminus, called the T-box to activate gene transcription when bound to such a site.

[00277] The disclosure also provides vaccines comprising Brachyury, CEA, or a combination thereof. In some embodiments, the antigen combination is contained in one vector as provided herein. In some embodiments, the antigen combination is contained in a separate vector as provided herein. [00278] In particular embodiments, there is provided a replication defective adenovirus vector of serotype 5 comprising a sequence encoding an immunogenic polypeptide. The immunogenic polypeptide may be an isoform of Brachyury or a subunit or a fragment thereof. In some

embodiments, the replication defective adenovirus vector comprises a sequence encoding a polypeptide with at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to the immunogenic polypeptide. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the following sequence identified by SEQ. ID. NO.:7. In some embodiments, the replication defective adenovirus vector comprises a sequence encoding a polypeptide with at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to the immunogenic polypeptide. In some embodiments, the sequence encoding the immunogenic polypeptide comprises the sequence of SEQ. ID. NO.: 8. In some embodiments, the sequence encoding the immunogenic polypeptide comprises a sequence with at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% identity to SEQ. ID. NO: 7, SEQ. ID. NO: 8 or a sequence generated from SEQ. ID. NO: 7, SEQ. ID. NO: 8 by alternative codon replacements. In some embodiments, the immunogenic polypeptide encoded by the adenovirus vectors described herein comprising up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, or more point mutations, such as single amino acid substitutions or deletions, as compared to a wild-type human Brachyury sequence.

[00279] In certain embodiments, the Brachyury antigen used herein is a wild-type antigen or a modified antigen. In certain embodiments, the Brachyury antigen binds to HLA-A2. In further embodiments, the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence set forth in WLLPGTSTV (SEQ ID NO:22), a HLA-A2 epitope of Brachyury. In further embodiments, the Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%), 100%) identity to SEQ ID NO:21, a modified Brachyury protein sequence. In certain embodiments, the replication-defective vector has a nucleotide sequence at least 80% identical SEQ ID NO:8 or positions 1033 to 2283 of SEQ ID NO: 16. In further embodiments, the second replication-defective vector has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to any portion or full-length of SEQ ID NO: 16 (the predicted sequence of an adenovirus vector express a modified Brachyury antigen), such as positions 1033 to 2283 of SEQ ID NO: 16. In some embodiments, the

Brachyury antigen is a modified Brachyury antigen having an amino acid sequence at least 80% identical to SEQ ID NO: 15 (another mutated Brachyury protein sequence). In certain embodiments, the second replication-defective vector or a replication-defective vector that express Brachyury has a nucleotide sequence at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to positions 520-1824 of SEQ ID

NO:7(wild-type Brachyury), SEQ ID NO:7 , or SEQ ID NO:8. Infectious Disease Associated Antigen Targets

[00280] Target antigens include, but are not limited to, antigens derived from any of a variety of infectious agents such as parasites, bacteria, virus, prions, and the like. An infectious agent may refer to any living organism capable of infecting a host. Infectious agents include, for example, bacteria, any variety of viruses, such as, single stranded RNA viruses, single stranded DNA viruses, fungi, parasites, and protozoa.

[00281] Examples of infectious disease associated target antigens that can be used with the compositions and the methods can be derived from the following: Actinobacillus spp., Actinomyces spp., Adenovirus (types 1, 2, 3, 4, 5 et 7), Adenovirus (types 40 and 41), Aerococcus spp.,

Aeromonas hydrophila, Ancylostoma duodenale, Angiostrongylus cantonensis, Ascaris

lumbricoides, Ascaris spp., Aspergillus spp., Babesia spp, B. microti, Bacillus anthracis, Bacillus cereus, Bacteroides spp., Balantidium coli, Bartonella bacilliformis, Blastomyces dermatitidis, Bluetongue virus, Bordetella bronchiseptica, Bordetella pertussis, Borrelia afzelii, Borrelia burgdorferi, Borrelia garinii, Branhamella catarrhalis, Brucella spp. (B. abortus, B. canis, B.

melitensis, B. suis), Brugia spp., Burkholderia, (Pseudomonas) mallei, Burkholderia (Pseudomonas) pseudomallei, California serogroup, Campylobacter fetus subsp. Fetus, Campylobacter jejuni, C. coli, C. fetus subsp. Jejuni, Candida albicans, Capnocytophaga spp., Chikungunya virus, Chlamydia psittaci, Chlamydia trachomatis, Citrobacter spp., Clonorchis sinensis, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Clostridium spp. (with the exception of those species listed above), Coccidioides immitis, Colorado tick fever virus,

Corynebacterium diphtheriae, Coxiella burnetii, Coxsackievirus, Creutzfeldt-Jakob agent, Kuru agent, Crimean-Congo hemorrhagic fever virus, Cryptococcus neoformans, Cryptosporidium parvum, Cytomegalovirus, Cyclospora cayatanesis, Dengue virus (1, 2, 3, 4), Diphtheroids, Eastern (Western) equine encephalitis virus, Ebola virus, Echinococcus granulosus, Echinococcus multilocularis, Echovirus, Edwardsiella tarda, Entamoeba histolytica, Enterobacter spp., Enterovirus 70, Epidermophyton floccosum, Ehrlichia spp, Ehrlichia sennetsu, Microsporum spp. Trichophyton spp., Epstein -Barr virus, Escherichia coli, enterohemorrhagic, Escherichia coli, enteroinvasive, Escherichia coli, enteropathogenic, Escherichia coli, enterotoxigenic, Fasciola hepatica, Francisella tularensis, Fusobacterium spp., Gemella haemolysans, Giardia lamblia, Guanarito virus,

Haemophilus ducreyi, Haemophilus influenzae (group b), Hantavirus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Hepatitis D virus, Hepatitis E virus, Herpes simplex virus, Herpesvirus simiae, Histoplasma capsulatum, Human coronavirus, Human immunodeficiency virus, Human papillomavirus, Human rotavirus, Human T-lymphotrophic virus, Influenza virus including H5N1, Junin virus/Machupo virus, Klebsiella spp., Kyasanur Forest disease virus, Lactobacillus spp., Lassa virus, Legionella pneumophila, Leishmania major, Leishmania infantum, Leishmania spp.,

Leptospira interrogans, Listeria monocytogenes, Lymphocytic choriomeningitis virus, Machupo virus, Marburg virus, Measles virus, Micrococcus spp., Moraxella spp., Mycobacterium spp. (other than M. bovis, M. tuberculosis, M. avium, M. leprae), Mycobacterium tuberculosis, M. bovis, Mycoplasma hominis, M. orale, M. salivarium, M. fermentans, Mycoplasma pneumoniae, Naegleria fowleri, Necator americanus, Neisseria gonorrhoeae, Neisseria meningitides, Neisseria spp. (other than N. gonorrhoeae and N. meningitidis), Nocardia spp., Norwalk virus, Omsk hemorrhagic fever virus, Onchocerca volvulus, Opisthorchis spp., Parvovirus B19, Pasteurella spp., Peptococcus spp., Peptostreptococcus spp., Plasmodium falciparum, Plasmodium vivax, Plasmodium spp.,

Plesiomonas shigelloides, Powassan encephalitis virus, Proteus spp., Pseudomonas spp. (other than P. mallei, P. pseudomallei), Rabies virus, Respiratory syncytial virus, Rhinovirus, Rickettsia akari, Rickettsia prowazekii, R. Canada, Rickettsia rickettsii, Rift Valley virus, Ross river virus/O'Nyong- Nyong virus, Rubella virus, Salmonella choleraesuis, Salmonella paratyphi, Salmonella typhi, Salmonella spp. (with the exception of those species listed above), Schistosoma spp., Scrapie agent, Serratia spp., Shigella spp., Sindbis virus, Sporothrix schenckii, St. Louis encephalitis virus, Murray Valley encephalitis virus, Staphylococcus aureus, Streptobacillus moniliformis, Streptococcus agalactiae, Streptococcus faecalis, Streptococcus pneumoniae, Streptococcus pyogenes,

Streptococcus salivarius, Taenia saginata, Taenia solium, Toxocara canis, T. cati, T. cruzi,

Toxoplasma gondii, Treponema pallidum, Trichinella spp., Trichomonas vaginalis, Trichuris trichiura, Trypanosoma brucei, Trypanosoma cruzi, Ureaplasma urealyticum, Vaccinia virus, Varicella-zoster virus, eastern equine encephalitis virus (EEEV), severe acute respiratory virus (SARS), Venezuelan equine encephalitis virus (VEEV), Vesicular stomatitis virus, Vibrio cholerae, serovar 01, Vibrio parahaemolyticus, West Nile virus, Wuchereria bancrofti, Yellow fever virus, Yersinia enterocolitica, Yersinia pseudotuberculosis, and Yersinia pestis. Target antigens may include proteins, or variants or fragments thereof, produced by any of the infectious organisms.

[00282] A number of viruses are associated with viral hemorrhagic fever, including filoviruses (e.g., Ebola, Marburg, and Reston), arenaviruses (e.g. Lassa, Junin, and Machupo), and

bunyaviruses. In addition, phleboviruses, including, for example, Rift Valley fever virus, have been identified as etiologic agents of viral hemorrhagic fever. Etiological agents of hemorrhagic fever and associated inflammation may also include paramyxoviruses, particularly respiratory syncytial virus. In addition, other viruses causing hemorrhagic fevers in man have been identified as belonging to the following virus groups: togavirus (Chikungunya), flavivirus (dengue, yellow fever, Kyasanur Forest disease, Omsk hemorrhagic fever), nairovirus (Crimian-Congo hemorrhagic fever) and hantavirus (hemorrhagic fever with renal syndrome, nephropathic epidemia). Furthermore, Sin Nombre virus was identified as the etiologic agent of the 1993 outbreak of hantavirus pulmonary syndrome in the American Southwest.

[00283] Target antigens may include viral coat proteins, i.e., influenza neuraminidase and hemagglutinin, HTV gpl60 or derivatives thereof, HTV Gag, HTV Nef, HTV Pol, SARS coat proteins, herpes virion proteins, W V proteins, etc. Target antigens may also include bacterial surface proteins including pneumococcal PsaA, PspA, LytA, surface or virulence associated proteins of bacterial pathogens such as Nisseria gonnorhea, outer membrane proteins or surface proteases.

HPV Antigen Targets

[00284] Target antigens may also include proteins, or variants or fragments thereof, associated with human papillomavirus (HPV), such as oncoproteins E6 and E7. In certain embodiments, the oncoprotein is modified to produce a non-oncogenic variant or a variant having reduced

oncogenicity relative to the wild type protein. For example, the portion of the peptide that is responsible for binding a tumor suppressor protein (e.g., p53 and pRb) may be deleted or modified so that it no longer interacts with the tumor suppressor protein. As another example, an oncoprotein that is a kinase, such as Her2/neu, may be kinase -inactivated, e.g., by point mutation. In some instances, two or more target antigens may be used during immunization. For example, the E6 and E7 antigens can be combined in a fusion protein, or separate vectors containing heterologous nucleotides encoding the modified or unmodified E6 and E7 target antigens are used in combination. For example, an Ad5-E6 vector can be administered with an Ad5-E7 vector. In this example, the Ad5-E6 vector and Ad5-E7 vector may be administered simultaneously or they may be administered sequentially.

[00285] High-risk human papillomavirus (HPV) such as HPV type- 16 is associated with the etiology of cervical and more than 90% of HPV-related head and neck squamous cell carcinomas. Preventive vaccines such as human papillomavirus bivalent [Types 16 and 18] vaccine and recombinant and human papillomavirus quadrivalent [Types 6, 11, 16, and 18] vaccine can be a primary defense against HPV associated cancers by preventing infection with the virus but reports indicate that they are not effective for active immunotherapy of established disease. The HPV early 6 (E6) and early 7 (E7) genes are expressed at high levels in HPV-induced cancers and are involved in the immortalization of primary human epidermal cells. Thus, these are ideal targets for tumor- specific immunotherapy because unlike many other tumor-associated antigens these viral antigens are "non-self and thus do not have the potential to induce autoimmunity.

[00286] Disclosed herein is an immunotherapy against human papilloma virus (HPV) using a viral gene delivery platform to immunize against HPV 16 genes E6 and E7 (Ad5 [E1 -, E2b-]-E6/E7) combined with programmed death-ligand 1 (PD1) blockade. Also disclosed herein is an

immunotherapeutic comprised of a gene delivery vehicle (Ad5 [E1 -, E2b-]) carrying modified genes for HPV type- 16 E6 and E7. The HPV E6 and E7 genes can be modified to render them non- oncogenic while retaining the antigenicity necessary to produce an immune response against HPV induced tumors. The modified genes can lack the capacity to degrade p53 , pRb, and PTPN13. The modified genes can be incorporated into a vaccine (Ad5 [E1 -, E2b-]-E6/E7). The Ad5 [E1 -, E2b-]- E6/E7 vaccine can retain the ability to induce an HPV-specific cell-mediated immune (CMI) response and can synergize with standard clinical therapy, enhancing immune-mediated clearance of an HPV-E6/E7 expressing tumor.

[00287] A balance between activation and inhibitory signals regulates the interaction between T lymphocytes and tumor cells, wherein T cell responses are initiated through antigen recognition by T-cell receptors (TCRs). In some cases, when combined with chemotherapy/radiation treatment in HPV-E6/E7 expressing tumor bearing mice, immunotherapy treatment with Ad5 [E1 -, E2b-]-E6/E7 can result in significant improvement in overall survival as compared to subjects that receive chemotherapy/radiation alone.

[00288] In particular embodiments, the HPV antigen used herein is a wild-type HPV antigen or a modified HPV antigen. For example, the modified HPV antigen a non-oncogenic HPV antigen or a modified HPV antigen that has reduced oncogenicity as compared with a wild-type HPV. For example, the antigen used herein is a modified HPV E6 antigen having an amino acid sequence set forth in SEQ ID NO: 17, a modified HPV E7 antigen having an amino acid sequence set forth in SEQ ID NO: 18, or a combination thereof. In further embodiments, the antigen used herein is a modified HPV E6 antigen that has one or more mutations at positions 26, 98, or 106 of SEQ ID NO: 17, a modified HPV E7 antigen that a mutation at position 86 of SEQ ID NO: 18, or a combination thereof. In particular embodiments, the nucleotide sequence of the antigen has a region at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identical to positions 123-545 of SEQ ID NO: 19, positions 602-895 of SEQ ID NO:9, or a combination thereof. For example, the nucleic acid sequence has at least 80% identity to SEQ ID NO: 19 (the nucleotide sequence of a HPV E6 and E7 fusion protein). In further embodiments, the nucleic acid sequence has at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9%, 100% identity to any portion of or full-length to SEQ ID NO:20 (the predicted sequence of an adenovirus vector expressing HPV E6 and E7), such as positions 1033 to 2179 of SEQ ID NO:20.

Personalized Tumor Associated Antigens

[00289] In certain embodiments tumor associated antigens used with the compositions and methods as described herein may be identified directly from an individual with a proliferative disease or cancer. In certain embodiments, cancers may include benign tumors, metastatic tumors, carcinomas, or sarcomas and the like. In some embodiments, a personalized tumor antigen comprises MUC1, MUClc, MUCln, T, or CEA characterized from a patient and further utilized as the target antigen as a whole, in part or as a variant.

[00290] In this regard, screens can be carried out using a variety of known technologies to identify tumor target antigens from an individual. For example, in one embodiment, a tumor biopsy is taken from a patient, RNA is isolated from the tumor cells and screened using a gene chip (for example, from Affymetrix, Santa Clara, Calif.) and a tumor antigen is identified. Once the tumor target antigen is identified, it may then be cloned, expressed and purified using techniques known in the art.

[00291] This target antigen can then linked to one or more epitopes or incorporated or linked to cassettes or viral vectors described herein and administered to the patient in order to alter the immune response to the target molecule isolated from the tumor. In this manner, "personalized" immunotherapy and vaccines are contemplated in certain embodiments. Where caner is genetic, that is inherited, for example the patient has been identified to have a BRAC1 or BRAC2 mutation, the vaccine can be used prophylactically. When the cancer is sporadic the immunotherapy can be used to reduce the size of the tumor, enhance overall and reduce reoccurrence of the cancer in a subject.

Combination Immunotherapies and Vaccines

[00292] Certain embodiments provide a combination immunotherapy and vaccine compositions for the treatment of cancer and infectious diseases. In some aspects, combination immunotherapies and vaccines provided herein can comprise a multi-targeted immunotherapeutic approach against antigens associated with the development of cancer such as tumor associated antigen, (TAA) or antigens know to be involved in a particular infectious disease, such as infectious disease associated antigen (IDAA). In some aspects, combination immunotherapies and vaccines provided herein can comprise a multi-targeted antigen signature immunotherapeutic approach against antigens associated with the development of cancer or infectious disease. The compositions and methods, in various embodiments, provide viral based vectors expressing a variant of MUC1, MUClc, MUCln, T, and/or CEA for immunization of a disease, as provided herein. These vectors can raise an immune response against MUC1, MUClc, MUCln, T, and/or CEA.

[00293] In some aspects, the vector comprises at least one antigen. In some aspects, the vector comprises at least two antigens. In some aspects, the vector comprises at least three antigens. In some aspects, the vector comprises more than three antigens. In some aspects, the vaccine formulation comprises 1 : 1 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :2 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :3 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :4 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :5 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :6 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :7 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :8 ratio of vector to antigen. In some aspects, the vaccine comprises 1 :9 ratio of vector to antigen. In some aspects, the vaccine comprises 1 : 10 ratio of vector to antigen.

[00294] In some aspects, the vaccine is a combination vaccine, wherein the vaccine comprises at least two vectors each containing at least a single antigen. In some aspects the vaccine is a combination vaccine, wherein the vaccine comprises at least three vectors each containing at least a single antigen target. In some aspects the vaccine is a combination vaccine, wherein the vaccine comprises more than three vectors each containing at least a single antigen.

[00295] In some aspects, the vaccine is a combination vaccine, wherein the vaccine comprises at least two vectors, wherein a first vector of the at least two vectors comprises at least a single antigen and wherein a second vector of the at least two vectors comprises at least two antigens. In some aspects, the vaccine is a combination vaccine, wherein the vaccine comprises at least three vectors, wherein a first vector of the at least three vectors comprises at least a single antigen and wherein a second vector of the at least three vectors comprises at least two antigens. In some aspects, the vaccine is a combination vaccine, wherein the vaccine comprises three or more vectors, wherein a first vector of the three or more vectors comprises at least a single antigen and wherein a second vector of the three or more vectors comprises at least two antigens. In some aspects the vaccine is a combination vaccine, wherein the vaccine comprises more than three vectors each containing at least two antigens.

[00296] When a mixture of different antigens are simultaneously administered or expressed from a same or different vector in an individual, they may compete with one another. As a result the formulations comprising different concentration and ratios of expressed antigens in a combination immunotherapy or vaccine must be evaluated and tailored to the individual or group of individuals to ensure that effective and sustained immune responses occur after administration.

[00297] Composition that comprises multiple antigens can be present at various ratios. For example, formulations with more than vector can have various ratios. For example, immunotherapies or vaccines can have two different vectors in a stoichiometry of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1 :7, 1:8, 1:9, 1:10, 1:15, 1:20, 1:30, 2:1, 2:3, 2:4, 2:5, 2:6, 2:7, 2:8, 3:1, 3:3, 3:4, 3:5, 3:6, 3:7, 3:8, 3: 1, 3:3, 3:4, 3:5, 3:6, 3:7, 3:8, 4: 1, 4:3, 4:5, 4:6, 4:7, 4:8, 5: 1, 5:3, 5:4, 5:6, 5:7, 5:8, 6:1, 6:3, 6:4, 6:5, 6:7, 6:8, 7: 1, 7:3, 7:4, 7:5, 7:6, 7:8, 8: 1, 8:3, 8:4, 8:5, 8:6, or 8:7. For example, immunotherapies or vaccines can have three different vectors in a stoichiometry of: 1:1:1, 1:2:1, 1:3:1, 1:4:1, 1:5:1, 1:6:1, 1:7:1, 1:8:1,2:1:1,2:3:1,2:4:1,2:5:1,2:6:1,2:7:1,2:8:1,3:1,3:3:1,3: 4:1,3:5:1,3:6:1,3:7:1,3:8:1, 3:1:1,3:3:1,3:4:1,3:5:1,3:6:1,3:7:1,3:8:1,4:1:1,4:3:1,4:4:1, 4:5:1,4:6:1,4:7:1,4:8:1,5:1:1, 5:3:1, 5:4:1, 5:5:1, 5:6:1, 5:7:1, 5:8:1, 6:1:1, 6:3:1, 6:4:1, 6:5:1, 6:6:1, 6:7:1, 6:8:1, 7:1:1, 7:3:1, 7:4:1, 7:5:1, 7:6:1, 7:7:1, 7:8:1, 8:1:1, 8:3:1, 8:4:1, 8:5:1, 8:6:1, 8:7:1, 8:8:1, 1:1:2, 1:2:2, 1:3:2, 1:4:2, 1:5:2, 1:6:2, 1:7:2, 1:8:2, 2:1:2, 2:3:2, 2:4:2, 2:5:2, 2:6:2, 2:7:2, 2:8:2, 3:1:2, 3:3:2, 3:4:2, 3:5:2, 3:6:2, 3:7:2, 3:8:2, 3:1:2, 3:3:2, 3:4:2, 3:5:2, 3:6:2, 3:7:2, 3:8:2, 4:1:2, 4:3:2, 4:4:2, 4:5:2, 4:6:2, 4:7:2, 4:8:2, 5:1:2, 5:3:2, 5:4:2, 5:5:2, 5:6:2, 5:7:2, 5:8:2, 6:1:2, 6:3:2, 6:4:2, 6:5:2, 6:6:2, 6:7:2, 6:8:2, 7:1:2, 7:3:2, 7:4:2, 7:5:2, 7:6:2, 7:7:2, 7:8:2, 8:1:2, 8:3:2, 8:4:2, 8:5:2, 8:6:2, 8:7:2, 8:8:2, 1:1:3, 1:2:3, 1:3:3, 1:4:3, 1:5:3, 1:6:3, 1:7:3, 1:8:3, 2:1:3, 2:3:3, 2:4:3, 2:5:3, 2:6:3, 2:7:3, 2:8:3, 3:1:3, 3:3:3, 3:4:3, 3:5:3, 3:6:3, 3:7:3, 3:8:3, 3:1:3, 3:3:3, 3:4:3, 3:5:3, 3:6:3, 3:7:3, 3:8:3, 4:1:3, 4:3:3, 4:4:3, 4:5:3, 4:6:3, 4:7:3, 4:8:3, 5:1:3, 5:3:3, 5:4:3, 5:5:3, 5:6:3, 5:7:3, 5:8:3, 6:1:3, 6:3:3, 6:4:3, 6:5:3, 6:6:3, 6:7:3, 6:8:3, 7:1:3, 7:3:3, 7:4:3, 7:5:3, 7:6:3, 7:7:3, 7:8:3, 8:1:3, 8:3:3, 8:4:3, 8:5:3, 8:6:3, 8:7:3, 8:8:3, 1:1:4, 1:2:4, 1:3:4, 1:4:4, 1:5:4, 1:6:4, 1:7:4, 1:8:4, 2:1:4, 2:3:4, 2:4:4, 2:5:4, 2:6:4, 2:7:4, 2:8:4, 3:1:4, 3:3:4, 3:4:4, 3:5:4, 3:6:4, 3:7:4, 3:8:4, 3:1:4, 3:3:4, 3:4:4, 3:5:4, 3:6:4, 3:7:4, 3:8:4, 4:1:4, 4:3:4, 4:4:4, 4:5:4, 4:6:4, 4:7:4, 4:8:4, 5:1:4, 5:3:4, 5:4:4, 5:5:4, 5:6:4, 5:7:4, 5:8:4, 6:1:4, 6:3:4, 6:4:4, 6:5:4, 6:6:4, 6:7:4, 6:8:4, 7:1:4, 7:3:4, 7:4:4, 7:5:4, 7:6:4, 7:7:4, 7:8:4, 8:1:4, 8:3:4, 8:4:3, 8:5:4, 8:6:4, 8:7:4, 8:8:4, 1:1:5, 1:2:5, 1:3:5, 1:4:5, 1:5:5, 1:6:5, 1:7:5, 1:8:5, 2:1:5, 2:3:5, 2:4:5, 2:5:5, 2:6:5, 2:7:5, 2:8:5, 3:1:5, 3:3:5, 3:4:5, 3:5:5, 3:6:5, 3:7:5, 3:8:5, 3:1:5, 3:3:5, 3:4:5, 3:5:5, 3:6:5, 3:7:5, 3:8:5, 4:1:5, 4:3:5, 4:4:5, 4:5:5, 4:6:5, 4:7:5, 4:8:5, 5:1:5, 5:3:5, 5:4:5, 5:5:5, 5:6:5, 5:7:5, 5:8:5, 6:1:5, 6:3:5, 6:4:5, 6:5:5, 6:6:5, 6:7:5, 6:8:5, 7:1:5, 7:3:5, 7:4:5, 7:5:5, 7:6:5, 7:7:5, 7:8:5, 8:1:5, 8:3:5, 8:4:5, 8:5:5, 8:6:5, 8:7:5, 8:8:5, 1:1:6, 1:2:6, 1:3:6, 1:4:6, 1:5:6, 1:6:6, 1:7:6, 1:8:6, 2:1:6, 2:3:6, 2:4:6, 2:5:6, 2:6:6, 2:7:6, 2:8:6, 3:1:6, 3:3:6, 3:4:6, 3:5:6, 3:6:6, 3:7:6, 3:8:6, 3:1:6, 3:3:6, 3:4:6, 3:5:6, 3:6:6, 3:7:6, 3:8:6, 4:1:6, 4:3:6, 4:4:6, 4:5:6, 4:6:6, 4:7:6, 4:8:6, 5:1:6, 5:3:6, 5:4:6, 5:5:6, 5:6:6, 5:7:6, 5:8:6, 6:1:6, 6:3:6, 6:4:6, 6:5:6, 6:6:6, 6:7:6, 6:8:6, 7:1:6, 7:3:6, 7:4:6, 7:5:6, 7:6:6, 7:7:6, 7:8:6, 8:1:6, 8:3:6, 8:4:6, 8:5:6, 8:6:5, 8:7:6, 8:8:6, 1:1:7, 1:2:7, 1:3:7, 1:4:7, 1:5:7, 1:6:7, 1:7:7, 1:8:7, 2:1:7, 2:3:7, 2:4:7, 2:5:7, 2:6:7, 2:7:7, 2:8:7, 3:1:7, 3:3:7, 3:4:7, 3:5:7, 3:6:7, 3:7:7, 3:8:7, 3:1:7, 3:3:7, 3:4:7, 3:5:7, 3:6:7, 3:7:7, 3:8:7, 4:1:7, 4:3:7, 4:4:7, 4:5:7, 4:6:7, 4:7:7, 4:8:7, 5:1:7, 5:3:7, 5:4:7, 5:5:7, 5:6:7, 5:7:7, 5:8:7, 6:1:7, 6:3:7, 6:4:7, 6:5:7, 6:6:7, 6:7:7, 6:8:7, 7:1:7, 7:3:7, 7:4:7, 7:5:7, 7:6:7, 7:7:7, 7:8:7, 8:1:7, 8:3:7, 8:4:7, 8:5:7, 8:6:5, 8:7:7, or 8:8:7.

[00298] Certain embodiments provide combination immunotherapies comprising multi-targeted immunotherapeutic directed TAAs. Certain embodiments provide combination immunotherapies comprising multi-targeted immunotherapeutic directed to IDAAs.

[00299] Certain embodiments provide a combination immunotherapies or vaccines comprising: at least two, at least three, or more than three different target antigens comprising a sequence encoding a modified CEA, MUClc, and/or Brachyury. For example, a combination immunotherapy or vaccine can comprise at least two, at least three, or more than three different target antigens comprising a sequence encoding a modified CEA, MUClc, and/or Brachyury, wherein the modified CEA compnses a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, or 99.9% to SEQ ID NO:l or SEQ ID NO:2; wherein the modified MUClc comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, or 99.9% to SEQ ID NO: 5 or SEQ ID NO:6; and wherein the modified Brachyury comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, 99.5%, 99.9% to SEQ ID NO:7 or SEQ ID NO:8. In some embodiments, the modified CEA comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% or 100% SEQ ID NO:l or SEQ ID NO:2 and has a Asn->Asp substitution at position 610. [00300] Certain embodiments provide combination immunotherapies comprising multi-targeted immunotherapeutic directed to TAAs and molecular compositions comprising an immune pathway checkpoint modulator that targets at least one immune checkpoint protein of the immune inhibitory pathway. Certain embodiments provide combination immunotherapies comprising multi-targeted immunotherapeutic directed to IDAAs and molecular compositions comprising an immune pathway checkpoint modulator that targets at least one immune checkpoint protein of the immune inhibitory pathway. Certain embodiments provide a combination immunotherapies or vaccines comprising: at least two, at least three, or more than three different target antigens comprising a sequence encoding a modified CEA, MUClc, and/or Brachyury, and at least one molecular composition comprising an immune pathway checkpoint modulator. For example, a combination immunotherapy or vaccine can comprise at least two, at least three, or more than three different target antigens comprising a sequence encoding a modified CEA, MUClc, and/or Brachyury, wherein the modified CEA comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, or 99.9% to SEQ ID NO: l or SEQ ID NO:2; wherein the modified MUClc comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, or 99.9% to SEQ ID NO: 5 or SEQ ID NO:6; wherein the modified Brachyury comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, 99.5%, 99.9% to SEQ ID NO:7 or SEQ ID NO: 8, and at least one molecular composition comprising an immune pathway checkpoint modulator. In some embodiments, the modified CEA comprises a sequence with an identity value of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, 99.5%, 99.9% or 100% SEQ ID NO: 1 or SEQ ID NO: 2 and has a Asn->Asp substitution at position 610.

[00301] In some embodiments, the immune pathway checkpoint modulator targets CTLA4. In some embodiments, the immune pathway checkpoint modulator targets PD1. In some embodiments, the immune pathway checkpoint modulator targets PDL1. In some embodiments, the immune pathway checkpoint modulator targets LAG3. In some embodiments, the immune pathway checkpoint modulator targets B7-H3. In some embodiments, the immune pathway checkpoint modulator targets B7-H4. In some embodiments, the immune pathway checkpoint modulator targets ΤΓΜ3. In some embodiment, the immune pathway checkpoint modulator is a monoclonal or polyclonal antibody directed to PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3 (i.e., HAVcr2), GAL9, and A2aR.

[00302] In some embodiments, at least one of the recombinant nucleic acid vectors is a replication defective adenovirus vector that comprises a replication defective adenovirus 5 vector comprising a first identity value. In some embodiments, the replication defective adenovirus vector comprises a deletion in the E2b region. In some embodiments, the replication defective adenovirus vector further comprises a deletion in the El region. In some embodiments, the first identity value is at least 90%. In some embodiments, the first identity value is at least 95%. In some embodiments, the first identity value is at least 99%. In some embodiments, the first identity value is 100%. In some embodiments, the first identity value is at least 90%. In some embodiments, the first identity value is at least 95%. In some embodiments, the first identity value is at least 99%. In some embodiments, the first identity value is 100%. In some embodiments, the first identity value is at least 90%. In some embodiments, the first identity value is at least 95%. In some embodiments, the first identity value is at least 99%. In some embodiments, the first identity value is 100%.

[00303] In some embodiments, at least one of the recombinant nucleic acid vector comprises a sequence with at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:3. In some embodiments, the recombinant nucleic acid vector comprises a region with at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to a region in SEQ ID NO:3, wherein the region is selected from 26048-26177, 26063-26141, 1-103, 54-103, 32214-32315, and 32214-32262. In some embodiments, the recombinant nucleic acid vector further comprises a region encoding a peptide with at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to a peptide encoded by a region in SEQ ID NO:3 between positions 1057 and 3165.

Combination Therapy

[00304] In certain embodiments, there is provided a method of treating a CEA-expression cancer in an individual in need thereof, the method comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective vector comprising a nucleic acid sequence encoding a CEA antigen or any suitable antigen; and administering to the individual an immune checkpoint inhibitor. The method may further comprise administering to the individual a VEGF inhibitor, a chemotherapy, or a combination thereof.

[00305] In certain embodiments, the chemotherapy used herein is capecitabine, leucovorin, fluorouracil, oxaliplatin, fluoropyrimidine, irinotecan, mitomycin, regorafenib, cetuxinab, panitumumab, acetinophen, or a combination thereof. In particular embodiments, the chemotherapy used herein is FOLFOX (leucovorin, fluorouracil and oxaliplatin) or capecitabine. In certain embodiments, the immune checkpoint inhibitor is an anti-PD-1 or anti-PD-Ll antibody, such as avelumab. In certain embodiments, the VEGF inhibitor is an anti-VEGF antibody, such as bevacizumab.

[00306] FOLFOX (5-fluorouracil, leucovorin, oxaliplatin)

[00307] A randomized trial comparing irinotecan and bolus fluorouracil plus leucovorin (IFL, control combination), oxaliplatin and infused fluorouracil plus leucovorin (FOLFOX), or irinotecan and oxaliplatin (IROX) established the FOLFOX combination, given for a total of 6 months, as the standard of care for first line treatment in patients with metastatic colorectal cancer (mCRC). Though multiple infusion schedules of FOLFOX have been validated, typically denominated as 'modified FOLFOX, there are no essential changes in the constituent cytotoxic agents of the regimen. Of these, mFOLFOX6 is one of the most widely used.

[00308] Oxaliplatin, however, is very difficult for patients to receive for greater than 6 months (12 cycles) due to progressive neurotoxicity. Though 6 months of combination therapy remains the standard of care in mCRC, clinical judgment may influence the decision to limit the number of oxaliplatin-containing cycles towards the end of treatment Other trials, including the CAIR03 study, have demonstrated the feasibility and benefit of discontinuation of oxaliplatin after a 3 month "induction" period with continuation of 5-FU and leucovorin as "maintenance" therapy.

[00309] Bevacizumab (Avastin®)

[00310] Addition of bevacizumab to first-line 5-FU and Oxaliplatin containing regimens was demonstrated to increase time to progression in mCRC patients with a manageable side effect profile and non-overlapping toxicities. Later trials indicated that continuing bevacizumab beyond first progression (in combination with subsequent chemotherapy) improved overall survival in an unselected group of patients by KRAS mutational status, which has led to its approved use in the maintenance setting.

[00311] Capecitabine

[00312] This agent is a prodrug that is enzymatically converted to 5-fluorouracil by 3 enzymatic steps following oral ingestion. As an orally active fluoropyrimidine, capecitabine has been approved for use in the adjuvant setting. In the advanced colon cancer setting, it has been shown to be equally efficacious as 5-fluorouracil, though with more reported rates of hand-foot syndrome. This agent offers the convenience of the oral route with its benefits of reducing infusion commitments for patients in the maintenance setting, while achieving high

concentrations intratumorally, given the higher concentrations of thymidine phosphorylase in tumor as compared to normal tissues.

Immunological Fusion Partner Antigen Targets

[00313] The viral vectors may also include nucleic acid sequences that encode proteins that increase the immunogenicity of the target antigen. In this regard, the protein produced following immunization with the viral vector containing such a protein may be a fusion protein comprising the target antigen of interest fused to a protein that increases the immunogenicity of the target antigen of interest.

[00314] In one embodiment, such an immunological fusion partner is derived from a

Mycobacterium sp., such as a Mycobacterium tuberculosis-derived Ral2 fragment. Ral2 compositions and methods for their use in enhancing the expression and/or immunogenicity of heterologous polynucleotide/polypeptide sequences are described in U.S. Patent Application 60/158,585 and U.S. Patent No. 7,009,042, which are herein incorporated by reference in their entirety. Briefly, Ral2 refers to a polynucleotide region that is a subsequence of a Mycobacterium tuberculosis MTB32A nucleic acid. MTB32A is a serine protease of 32 kDa encoded by a gene in virulent and avirulent strains of M. tuberculosis. The nucleotide sequence and amino acid sequence of MTB32A have been described (see, e.g., U.S. Patent Application 60/158,585; Skeiky et al., Infection and Immun. 67:3998-4007 (1999), incorporated herein by reference in their entirety). C- terminal fragments of the MTB32A coding sequence express at high levels and remain as soluble polypeptides throughout the purification process. Moreover, Ral2 may enhance the immunogenicity of heterologous immunogenic polypeptides with which it is fused. One Ral2 fusion polypeptide comprises a 14 kDa C-terminal fragment corresponding to amino acid residues 192 to 323 of MTB32A. Other Ral2 polynucleotides generally comprise at least about 15, 30, 60, 100, 200, 300, or more nucleotides that encode a portion of a Ral2 polypeptide. Ral2 polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a Ral2 polypeptide or a portion thereof) or may comprise a variant of such a sequence. Ral2 polynucleotide variants may contain one or more substitutions, additions, deletions and/or insertions such that the biological activity of the encoded fusion polypeptide is not substantially diminished, relative to a fusion polypeptide comprising a native Ral2 polypeptide. Variants can have at least about 70%, 80%, or 90% identity, or more, to a polynucleotide sequence that encodes a native Ral2 polypeptide or a portion thereof.

[00315] An immunological fusion partner can be derived from protein D, a surface protein of the gram-negative bacterium Haemophilus influenza B. In some cases, a protein D derivative comprises approximately the first third of the protein (e.g., the first N-terminal 100-110 amino acids). A protein D derivative may be lipidated. Within certain embodiments, the first 109 residues of a Lipoprotein D fusion partner is included on the N-terminus to provide the polypeptide with additional exogenous T- cell epitopes, which may increase the expression level in E. coli and may function as an expression enhancer. The lipid tail may ensure optimal presentation of the antigen to antigen presenting cells. Other fusion partners include the non-structural protein from influenza virus, NS1 (hemagglutinin). Typically, the N-terminal 81 amino acids are used, although different fragments that include T- helper epitopes may be used.

[00316] The immunological fusion partner can be the protein known as LYTA, or a portion thereof (preferably a C-terminal portion). LYTA is derived from Streptococcus pneumoniae, which synthesizes an N-acetyl-L-alanine amidase known as amidase LYTA (encoded by the LytA gene). LYTA is an autolysin that specifically degrades certain bonds in the peptidoglycan backbone. The C-terminal domain of the LYTA protein is responsible for the affinity to the choline or to some choline analogues such as DEAE. This property has been exploited for the development of E. coli C- LYTA expressing plasmids useful for expression of fusion proteins. Purification of hybrid proteins containing the C-LYTA fragment at the amino terminus can be employed. Within another embodiment, a repeat portion of LYTA may be incorporated into a fusion polypeptide. A repeat portion can, for example, be found in the C-terminal region starting at residue 178. One particular repeat portion incorporates residues 188-305.

[00317] In some embodiments, the antigen target comprises an immunogenic component comprising a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13. In some embodiments, the antigen target further comprises one or more immunogenic component comprises a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13. In some embodiments, the antigen target comprises an immunogenic component comprising a nucleic acid encoding of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13., a protein with substantial identity to one or more of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13, and a nucleic acid encoding a protein with substantial identity to one or more of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13.

[00318] In some embodiments, the antigen target is fused or linked to an immunogenic component comprising a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13. In some embodiments, the antigen target is co-expressed in a cell with an immunogenic component comprising a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13.

Immune Pathway Checkpoint Modulators

[00319] In some embodiments, compositions are administered with one or more immune checkpoint modulator, such as immune checkpoint inhibitors. In some embodiments, the

composition comprises a replication-defective vector comprising a nucleotide sequence encoding a target antigen, such as CEA, MUCl, Brachyury, HPV E6, HPV E7, or a combination thereof, or any suitable antigens.

[00320] A balance between activation and inhibitory signals regulates the interaction between T lymphocytes and disease cells, wherein T-cell responses are initiated through antigen recognition by the T-cell receptor (TCR). The inhibitory pathways and signals are referred to as immune checkpoints. In normal circumstances, immune checkpoints play a critical role in control and prevention of autoimmunity and also protect from tissue damage in response to pathogenic infection.

[00321] In certain aspect, there are provided combination immunotherapies comprising viral vector based vaccines and compositions for modulating immune checkpoint inhibitory pathways for the treatment of cancer and infectious diseases. In some embodiments, modulating is increasing expression or activity of a gene or protein. In some embodiments, modulating is decreasing expression or activity of a gene or protein. In some embodiments, modulating affects a family of genes or proteins. [00322] In general, the immune inhibitory pathways are initiated by ligand-receptor interactions. It is now clear that in diseases, the disease can co-opt immune-checkpoint pathways as mechanism for inducing immune resistance in a subject.

[00323] The induction of immune resistance or immune inhibitory pathways in a subject by a given disease can be blocked by molecular compositions such as siRNAs, antisense, small molecules, mimic, a recombinant form of ligand, receptor or protein, or antibodies (which can be an Ig fusion protein) that are known to modulate one or more of the Immune Inhibitory Pathways. For example, preliminary clinical findings with blockers of immune-checkpoint proteins, such as Cytotoxic T- lymphocyte-associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) have shown promise for enhancing antitumor immunity.

[00324] Because diseased cells can express multiple inhibitory ligands, and disease-infiltrating lymphocytes express multiple inhibitory receptors, dual or triple blockade of immune checkpoints proteins may enhance anti-disease immunity. Combination immunotherapies as provide herein can comprise one or more molecular compositions of the following immune-checkpoint proteins: PD1 , PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3 (also known as CD276), B7-H4 (also known as B7-S1, B7x and VCTN1), BTLA (also known as CD272), HVEM, KIR, TCR, LAG3 (also known as CD223), CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3 (also known as HAVcr2), GAL9, and A2aR. In some embodiments, the molecular composition comprises siRNAs. In some embodiments, the molecular composition comprises a small molecule. In some embodiments, the molecular composition comprises a recombinant form of a ligand. In some embodiments, the molecular composition comprises a recombinant form of a receptor. In some embodiments, the molecular composition comprises an antibody. In some embodiments, the combination therapy comprises more than one molecular composition and/or more than one type of molecular composition. As it will be appreciated by those in the art, future discovered proteins of the immune checkpoint inhibitory pathways are also envisioned to be encompassed in certain aspects.

[00325] In some embodiments, combination immunotherapies comprise molecular compositions for the modulation of CTLA4. In some embodiments, combination immunotherapies comprise molecular compositions for the modulation PD1. In some embodiments, combination

immunotherapies comprise molecular compositions for the modulation PDL1. In some

embodiments, combination immunotherapies comprise molecular compositions for the modulation LAG3. In some embodiments, combination immunotherapies comprise molecular compositions for the modulation B7-H3. In some embodiments, combination immunotherapies comprise molecular compositions for the modulation B7-H4. In some embodiments, combination immunotherapies comprise molecular compositions for the modulation TIM3. In some embodiments, modulation is an increase or enhancement of expression. In other embodiments, modulation is the decrease of absence of expression.

[00326] Two exemplary immune checkpoint inhibitors include the cytotoxic T lymphocyte associated antigen-4 (CTLA-4) and the programmed cell death protein-1 (PDl). CTLA-4 can be expressed exclusively on T-cells where it regulates early stages of T-cell activation. CTLA-4 interacts with the co-stimulatory T-cell receptor CD28 which can result in signaling that inhibits T- cell activity. Once TCR antigen recognition occurs, CD28 signaling may enhances TCR signaling, in some cases leading to activated T-cells and CTLA-4 inhibits the signaling activity of CD28. Certain embodiments provide immunotherapies as provided herein in combination with anti-CTLA-4 monoclonal antibody for the treatment of proliferative disease and cancer. Certain embodiments provide immunotherapies as provided herein in combination with CTLA-4 molecular compositions for the treatment of proliferative disease and cancer.

[00327] Programmed death cell protein ligand-1 (PDLl) is a member of the B7 family and is distributed in various tissues and cell types. PDLl can interact with PDl inhibiting T-cell activation and CTL mediated lysis. Significant expression of PDLl has been demonstrated on various human tumors and PDLl expression is one of the key mechanisms in which tumors evade host antitumor immune responses. Programmed death-ligand 1 (PDLl) and programmed cell death protein-1 (PDl) interact as immune checkpoints. This interaction can be a major tolerance mechanism which results in the blunting of anti -tumor immune responses and subsequent tumor progression. PDl is present on activated T cells and PDLl, the primary ligand of PDl, is often expressed on tumor cells and antigen-presenting cells (APC) as well as other cells, including B cells. Significant expression of PDLl has been demonstrated on various human tumors including HPV-associated head and neck cancers. PDLl interacts with PDl on T cells inhibiting T cell activation and cytotoxic T lymphocyte (CTL) mediated lysis. Certain embodiments provide immunotherapies as provided herein in combination with anti-PDl or anti-PDLl monoclonal antibody for the treatment of proliferative disease and cancer. Certain embodiments provide immunotherapies as provided herein in combination with PDl or anti-PDLl molecular compositions for the treatment of proliferative disease and cancer. Certain embodiments provide immunotherapies as provided herein in combination with anti-CTLA-4 and anti-PDl monoclonal antibodies for the treatment of proliferative disease and cancer. Certain embodiments provide immunotherapies as provided herein in combination with anti-CTLA-4 and PDLl monoclonal antibodies for the treatment of proliferative disease and cancer. Certain embodiments provide immunotherapies as provided herein in combination with anti-CTLA-4, anti-PDl, PDLl, monoclonal antibodies, or a combination thereof, for the treatment of proliferative disease and cancer.

[00328] Certain embodiments provide immunotherapies as provided herein in combination with several antibodies directed against the PD-L1 / PD-1 pathway that are in clinical development for cancer treatment. In certain embodiments, anti-PD-Ll antibodies may be used. Compared with anti-PD-1 antibodies that target T-cells, anti-PDLl antibodies that target tumor cells are expected to have less side effects, including a lower risk of autoimmune-related safety issues, as blockade of PD-Ll leaves the PD-L2 / PD-1 pathway intact to promote peripheral self-tolerance.

[00329] To this end, avelumab, a fully human IgGl anti-PDLl antibody (drug code

MSB0010718C) has been produced. Avelumab selectively binds to PD-Ll and competitively blocks its interaction with PD-1.

[00330] Avelumab is also cross-reactive with murine PD-Ll, thus allowing in vivo

pharmacology studies to be conducted in normal laboratory mice. However, due to

immunogenicity directed against the fully human avelumab molecule, the dosing regimen was limited to three doses given within a week.

[00331] The key preclinical pharmacology findings for avelumab are summarized below.

Avelumab showed functional enhancement of primary T cell activation in vitro in response to antigen-specific and antigen non-specific stimuli; and significant inhibition of in vivo tumor growth (PD-Ll expressing MC38 colon carcinoma) as a monotherapy. Its in vivo efficacy is driven by CD8+ T cells, as evidenced by complete abrogation of anti-tumor activity when this cell type was systemically depleted. Its combination with localized, fractionated radiotherapy resulted in complete regression of established tumors with generation of anti-tumor immune memory. Its use in chemotherapy combinations also showed promising activity: additive combination effect when partnered with oxaliplatin and 5-fluorouracil (5-FU) (core components of FOLFOX [oxaliplatin, 5-FU, and folinic acid]) against MC38 colon tumors; significant increase in survival when partnered with gemcitabine against PANC02 pancreatic tumors. Its antibody-dependent cell-mediated cytotoxicity (ADCC) was demonstrated against human tumor cells in vitro; furthermore, studies in ADCC deficient settings in vivo support a contribution of ADCC to anti-tumor efficacy. Additional findings of Avelumab include: no complement- dependent cytotoxicity was observed in vitro. Immunomonitoring assays with translational relevance for the clinic further support an immunological mechanism of action: consistent increases in CD8+PD-1+ T cells and CD8+ effector memory T cells as measured by

fluorescence-activated cell sorter (FACS); enhanced tumor-antigen specific CD8+ T cell responses as measured by pentamer staining and enzyme-linked immunosorbent spot

(ELISPOT) assays.

[00332] Despite reports indicating that anti-tumor radiographic responses were unlikely using agents that interfere with PD-1— PD-Ll binding in colorectal cancer, there have been reports of radiographic responses. Additionally, a correlation has been demonstrated in multiple clinical trials indicating that PD-Ll expression levels on tumor tissue predict the likelihood of radiographic response. However, it has become clear that PD-L1 expression, as it is currently measured, is not a definitive requirement for anti-tumor efficacy. It has been noted that colorectal tumors rarely express PD-L1 compared with other tumors that are more likely to respond to PD-1 - PD-L1 blockade. However, it is known that a strong anti-tumor T cell response, producing IFN-gamma, will induce PD-L1 expression.

[00333] In some embodiments, without being bound by theory, it was contemplated that an underlying immune response is necessary for PD-1 - PD-L1 blockade to have an anti-tumor effect. Without being bound by theory, it was further contemplated that this combination of an immune checkpoint inhibitor with the standard therapy and an adenoviral vector composition such as Ad-CEA immunizations, Ad-E6/E7 immunizations, or Ad-CEA/MUCl/Brachyury immunizations may be capable of induction of PD-L1 expression and thereby increase the antitumor activity of PD-1 - PD-L1 blockade.

[00334] Some embodiments provide Ad5 [E1-, E2b-]-E6/E7 immunizations combined with PD1 blockade that can increase an anti -tumor effect. A CMI response induced by the Ad5 [E1-, E2b-]- E6/E7 vaccine was characterized to show kinetics of an anti-tumor response to evaluate the therapeutic potential of treating small versus large established tumors.

[00335] Immune checkpoint molecules can be expressed by T cells. Immune checkpoint molecules can effectively serve as "brakes" to down-modulate or inhibit an immune response. Immune checkpoint molecules include, but are not limited to Programmed Death 1 (PD1, also known as PDCDl or CD279, accession number: NM_005018), Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4, also known as CD152, GenBank accession number AF414120.1), LAG3 (also known as CD223, accession number: NM_002286.5), Tim3 (also known as HAVCR2, GenBank accession number: JX049979.1), BTLA (also known as CD272, accession number: NM_181780.3), BY55 (also known as CD160, GenBank accession number: CR541888.1), TIGIT (also known as IVSTM3, accession number: NM_173799), LAIR1 (also known as CD305, GenBank accession number: CR542051.1), SIGLECIO (GeneBank accession number: AY358337.1), 2B4 (also known as CD244, accession number: NM 001166664.1), PPP2CA, PPP2CB, PTPN6, PTPN22, CD96, CRTAM, SIGLEC7, SIGLEC9, TNFRSF10B, TNFRSF10A, CASP8, CASP10, CASP3, CASP6, CASP7, FADD, FAS, TGFBRII, TGFRBRI, SMAD2, SMAD3, SMAD4, SMADIO, SKI, SKIL, TGIFl, ILIORA, ILIORB, HMOX2, IL6R, IL6ST, EIF2AK4, CSK, PAG1, SIT1, FOXP3, PRDM1, BATF, GUCY1A2, GUCY1A3, GUCY1B2, GUCY1B3 which directly inhibit immune cells. For example, PD1 can be combined with an adenoviral vaccine to treat a patient in need thereof. Table 1, without being exhaustive, shows exemplary immune checkpoint genes that can be inactivated to improve the efficiency of the adenoviral vaccine. Immune checkpoints gene can be selected from such genes listed in Table 1 and others involved in co-inhibitory receptor function, cell death, cytokine signaling, arginine tryptophan starvation, TCR signaling, Induced T-reg repression, transcription factors controlling exhaustion or anergy, and hypoxia mediated tolerance.

Table 1

modulator may result in reduction in cancer recurrences in treated patients, as compared to either agent alone. In yet another embodiment the combination of an adenoviral-based vaccine and an immune pathway checkpoint modulator may result in reduction in the presence or appearance of metastases or micro metastases in treated patients, as compared to either agent alone. In another embodiment, the combination of a adenoviral-based vaccine and an immune pathway checkpoint modulator may result improved overall survival of treated patients, as compared to either agent alone. In some cases, the combination of an adenoviral vaccine and an immune pathway checkpoint modulator may increase the frequency or intensity of tumor-specific T cell responses in patients compared to either agent alone.

[00337] Some embodiments also disclose the use of immune checkpoint inhibition to improve performance of an adenoviral vector-based vaccine. The immune checkpoint inhibition may be administered at the time of the vaccine. The immune checkpoint inhibition may also be administered after a vaccine. Immune checkpoint inhibition may occur simultaneously to an adenoviral vaccine administration. Immune checkpoint inhibition may occur 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, or 60 minutes after vaccination. Immune checkpoint inhibition may also occur

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 post vaccination. In some cases, immune inhibition may occur 1, 2, 3, 4, 5, 6, or 7 days after vaccination. Immune checkpoint inhibition may occur at any time before or after vaccination.

[00338] In another aspect, there is provided a vaccine comprising an antigen and an immune pathway checkpoint modulator. Some embodiments pertain to a method for treating a subject having a condition that would benefit from downregulation of an immune checkpoint, PD1 for example, and its natural binding partner(s) on cells of the subject.

[00339] An immune pathway checkpoint modulator may be combined with an adenoviral vaccine comprising nucleotide sequences encoding any antigen. For example, an antigen can be MUClc, HER3, Brachyury, HER2NEU, CEA, or PSA. An immune pathway checkpoint modulator may produce a synergistic effect when combined with a vaccine. An immune pathway checkpoint modulator may also produce an additive effect when combined with a vaccine.

[00340] In particular embodiments, a checkpoint immune inhibitor may be combined with a vector comprising nucleotide sequences encoding any antigen, optionally with a chemotherapy or any other cancer care or therapy, such as VEGF inhibitors, angiogenesis inhibitors, radiation, other immune therapy, or any suitable cancer care or therapy.

Formulations

[00341] Some embodiments provide pharmaceutical compositions comprising a vaccination regime that can be administered either alone or together with a pharmaceutically acceptable carrier or excipient, by any routes, and such administration can be carried out in both single and multiple dosages. More particularly, the pharmaceutical composition can be combined with various pharmaceutically acceptable inert carriers in the form of tablets, capsules, lozenges, troches, hand candies, powders, sprays, aqueous suspensions, injectable solutions, elixirs, syrups, and the like. Such carriers include solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents, etc. Moreover, such oral pharmaceutical formulations can be suitably sweetened and/or flavored by means of various agents of the type commonly employed for such purposes. The compositions described throughout can be formulated into a pharmaceutical medicament and be used to treat a human or mammal, in need thereof, diagnosed with a disease, e.g., cancer.

[00342] For administration, viral vector stock can be combined with an appropriate buffer, physiologically acceptable carrier, excipient or the like. In certain embodiments, an appropriate number of virus vector particles (VP) are administered in an appropriate buffer, such as, sterile PBS or saline. In certain embodiment, vector compositions disclosed herein are provided in specific formulations for subcutaneously, parenterally, intravenously, intramuscularly, or even

intraperitoneally administration. In certain embodiments, formulations in a solution of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, squalene-based emulsion, Squalene-based oil-in-water emulsions, water-in-oil emulsions, oil-in-water emulsions, nonaqueous emulsions, water-in-paraffin oil emulsion, and mixtures thereof and in oils. In other embodiments, viral vectors may are provided in specific formulations for pill form administration by swallowing or by suppository. [00343] Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (see, e.g., U.S. Pat. No. 5,466,468). Fluid forms to the extent that easy syringability exists may be preferred. Forms that are stable under the conditions of manufacture and storage are provided in some embodiments. In various embodiments, forms are preserved against the contaminating action of microorganisms, such as bacteria, molds and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants. The prevention of the action of microorganisms can be facilitated by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and thimerosal. It may be suitable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

[00344] In one embodiment, for parenteral administration in an aqueous solution, the solution can be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 rriL of isotonic NaCl solution and either added to 1000 mL of hypodermoclysis fluid or injected at the proposed site of infusion, (see, e.g., "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage may occur depending on the condition of the subject being treated.

[00345] Carriers of formulation can comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.

[00346] In certain embodiments, the viral vectors may be administered in conjunction with one or more immunostimulants, such as an adjuvant. An immunostimulant refers to essentially any substance that enhances or potentiates an immune response (antibody and/or cell -mediated) to an antigen. One type of immunostimulant comprises an adjuvant. Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins. Certain adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories); Merck Adjuvant 65 (Merck and Company, Inc.) AS-2 (SmithKline Beecham); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides;

polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A. Cytokines, such as GM-CSF, IFN-γ, TNFa, IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and/or IL-13, and others, like growth factors, may also be used as adjuvants.

[00347] Within certain embodiments, the adjuvant composition can be one that induces an immune response predominantly of the Thl type. High levels of Thl -type cytokines (e.g., IFN-γ, TNFa, IL-2 and IL-12) tend to favor the induction of cell mediated immune responses to an administered antigen. In contrast, high levels of Th2-type cytokines (e.g., IL-4, IL-5, IL-6 and IL-10) tend to favor the induction of humoral immune responses. Following application of a vaccine as provided herein, a patient may support an immune response that includes Thl - and/or Th2-type responses. Within certain embodiments, in which a response is predominantly Thl -type, the level of Thl -type cytokines will increase to a greater extent than the level of Th2-type cytokines. The levels of these cytokines may be readily assessed using standard assays. Thus, various embodiments relate to therapies raising an immune response against a target antigen, for example MUC1, MUClc, MUCln, T, or CEA, using cytokines, e.g. IFN-γ, TNFa, IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and/or IL-13 supplied concurrently with a replication defective viral vector treatment. In some embodiments, a cytokine or a nucleic acid encoding a cytokine, is administered together with a replication defective viral described herein. In some embodiments, cytokine administration is performed prior or subsequent to viral vector administration. In some

embodiments, a replication defective viral vector capable of raising an immune response against a target antigen, for example MUC1, MUClc, MUCln, T, and/or CEA, further comprises a sequence encoding a cytokine.

[00348] Certain illustrative adjuvants for eliciting a predominantly Thl -type response include, for example, a combination of monophosphoryl lipid A, such as 3-de-O-acylated monophosphoryl lipid A, together with an aluminum salt. MPL® adjuvants are commercially available (see, e.g., U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094). CpG-containing oligonucleotides (in which the CpG dinucleotide is unmethylated) also induce a predominantly Thl response, (see, e.g. , WO 96/02555, WO 99/33488 and U.S. Pat. Nos. 6,008,200 and 5,856,462). Immunostimulatory DNA sequences can also be used. Another adjuvant for use comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc.), Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins. Other formulations may include more than one saponin in the adjuvant combinations, e.g. , combinations of at least two of the following group comprising QS21, QS7, Quil A, β-escin, or digitonin. [00349] In some embodiments, the compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles. The delivery of drugs using intranasal microparticle resins and lysophosphatidyl-glycerol compounds can be employed (see, e.g., U.S. Pat. No. 5,725,871). Likewise, illustrative transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix can be employed (see, e.g., U.S. Pat. No. 5,780,045).

[00350] Liposomes, nanocapsules, microparticles, lipid particles, vesicles, and the like, can be used for the introduction of the compositions into suitable hot cells/organisms. Compositions as described herein may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like. Alternatively, compositions as described herein can be bound, either covalently or non-covalently, to the surface of such carrier vehicles. Liposomes can be used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals. Furthermore, the use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery. In some embodiments, liposomes are formed from phospholipids dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (i.e. multilamellar vesicles (MLVs).

[00351] In some embodiments, pharmaceutically-acceptable nanocapsule formulations of the compositions are provided. Nanocapsules can generally entrap compounds in a stable and reproducible way. To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 μηι) may be designed using polymers able to be degraded in vivo.

[00352] The compositions in some embodiments comprise or are administered with a

chemotherapeutic agent (e.g., a chemical compound useful in the treatment of

cancer). Chemotherapeutic cancer agents that can be used in combination with the disclosed T cell include, but are not limited to, mitotic inhibitors (vinca alkaloids), such as vincristine, vinblastine, vindesine and Navelbine™ (vinorelbine,5'-noranhydroblastine); topoisomerase I inhibitors, such as camptothecin compounds (e.g., Camptosar™ (irinotecan HCL), Hycamtin™ (topotecan HCL) and other compounds derived from camptothecin and its analogues); podophyllotoxin derivatives, such as etoposide, teniposide and mitopodozide; alkylating agents such as cisplatin, cyclophosphamide, nitrogen mustard, trimethylene thiophosphoramide, carmustine, busulfan, chlorambucil, belustine, uracil mustard, chlomaphazin, and dacarbazine; antimetabolites such as cytosine arabinoside, fluorouracil, methotrexate, mercaptopurine, azathioprime, and procarbazine; antibiotics, such as doxorubicin, bleomycin, dactinomycin, daunorubicin, mithramycin, mitomycin, mytomycin C, and daunomycin; anti-tumor antibodies; dacarbazine; azacytidine; amsacrine; melphalan; ifosfamide; and mitoxantrone.

[00353] Compositions disclosed herein can be administered in combination with other anti-tumor agents, including cytotoxic/antineoplastic agents and anti -angiogenic agents. Cytotoxic/anti- neoplastic agents can be defined as agents who attack and kill cancer cells. Some cytotoxic/antineoplastic agents can be alkylating agents, which alkylate the genetic material in tumor cells, e.g., cis-platin, cyclophosphamide, nitrogen mustard, trimethylene thiophosphoramide, carmustine, busulfan, chlorambucil, belustine, uracil mustard, chlomaphazin, and dacabazine. Other

cytotoxic/anti-neoplastic agents can be antimetabolites for tumor cells, e.g., cytosine arabinoside, fluorouracil, methotrexate, mercaptopuirine, azathioprime, and procarbazine. Other cytotoxic/antineoplastic agents can be antibiotics, e.g., doxorubicin, bleomycin, dactinomycin, daunorubicin, mithramycin, mitomycin, mytomycin C, and daunomycin. There are numerous liposomal formulations commercially available for these compounds. Still other cytotoxic/anti-neoplastic agents can be mitotic inhibitors (vinca alkaloids). These include vincristine, vinblastine and etoposide. Miscellaneous cytotoxic/anti-neoplastic agents include taxol and its derivatives, L- asparaginase, anti-tumor antibodies, dacarbazine, azacytidine, amsacrine, melphalan, VM-26, ifosfamide, mitoxantrone, and vindesine.

[00354] Anti-angiogenic agents can also be used. Suitable anti-angiogenic agents for use in the disclosed methods and compositions include anti-VEGF antibodies, including humanized and chimeric antibodies, anti-VEGF aptamers and antisense oligonucleotides. Other inhibitors of angiogenesis include angiostatin, endostatin, interferons, interleukin 1 (including a and β) interleukin 12, retinoic acid, and tissue inhibitors of metalloproteinase-1 and -2. (TFMP-l and -2). Small molecules, including topoisomerases such as razoxane, a topoisomerase II inhibitor with anti- angiogenic activity, can also be used.

Methods of Preparation

[00355] In some embodiments, compositions and methods make use of human cytolytic T-cells (CTLs), such as those that recognize MUCl, T, or CEAs epitopes which bind to selected MHC molecules, e.g. HLA- A2, A3, and A24. Individuals expressing MHC molecules of certain serotypes, e.g. HLA- A2, A3, and A24 may be selected for therapy using the methods and compositions as described herein. For example, individuals expressing MHC molecules of certain serotypes, e.g. HLA- A2, A3, and A24, may be selected for a therapy including raising an immune response against MUCl, T, or CEAs, using the methods and compositions described herein.

[00356] In various embodiments, these T-cells can be generated by in vitro cultures using antigen- presenting cells pulsed with the epitope of interest to stimulate peripheral blood mononuclear cells. In addition, T-cell lines can also be generated after stimulation with MUCl, T, or CEAs latex beads, MUCl, T, or CEAs protein -pulsed plastic adherent peripheral blood mononuclear cells, or DCs sensitized with MUCl, T, or CEAs RNA. T-cells can also be generated from patients immunized with a vaccine vector encoding MUCl, T, or CEAs immunogen. HLA A2-presented peptides from MUCl, T, or CEAs can further be found in primary gastrointestinal tumors. [00357] Some embodiments relate to an HLA A2 restricted epitope of MUCl, T, or CEAs, CAP-1, a nine amino acid sequence (YLSGANLNL; SEQ. ID. NO.:4), with ability to stimulate CTLs from cancer patients immunized with vaccine-MUCl, T, or CEAs. Cap-1(6D) (YLSGADLNL; SEQ. ID. NO.: 10) is a peptide analog of CAP-1. Its sequence includes a heteroclitic (nonanchor position) mutation, resulting in an amino acid change from Asn to Asp, enhancing recognition by the T-cell receptor. The Asn to Asp mutation appears to not cause any change in the binding of the peptide to HLA A2. Compared with the non-mutated CAP-1 epitope, Cap-1(6D) can enhance the sensitization of CTLs by 100 to 1,000 times. CTL lines can be elicited from peripheral blood mononuclear cells of healthy volunteers by in vitro sensitization to the Cap-1(6D) peptide, but not significantly to the CAP-1 peptide. These cell lines can lyse human tumor cells expressing endogenous CEA. Thus, polypeptide sequences comprising CAP-1 or CAP-1(6D), nucleic acid sequences encoding such sequences, an adenovirus vectors; for example replication defective adenovirus vectors, comprising such nucleic acid sequences are provided in some embodiments.

Methods of Treatment

[00358] The adenovirus vectors can be used in a number of vaccine settings for generating an immune response against one or more target antigens as described herein. Some embodiments provide methods of generating an immune response against any target antigen, such as those described elsewhere herein. The adenovirus vectors are of particular importance because of the unexpected finding that they can be used to generate immune responses in subjects who have preexisting immunity to Ad and can be used in vaccination regimens that include multiple rounds of immunization using the adenovirus vectors, regimens not possible using previous generation adenovirus vectors.

[00359] Generally, generating an immune response comprises an induction of a humoral response and/or a cell-mediated response. It may desirable to increase an immune response against a target antigen of interest. Generating an immune response may involve a decrease in the activity and/or number of certain cells of the immune system or a decrease in the level and/or activity of certain cytokines or other effector molecules. Any suitable methods for detecting alterations in an immune response (e.g., cell numbers, cytokine expression, cell activity) can be used in some embodiments. Illustrative methods useful in this context include intracellular cytokine staining (ICS), ELISpot, proliferation assays, cytotoxic T-cell assays including chromium release or equivalent assays, and gene expression analysis using any number of polymerase chain reaction (PCR) or RT-PCR based assays.

[00360] Generating an immune response can comprise an increase in target antigen-specific CTL activity of between 1.5 and 5 fold in a subject administered the adenovirus vectors as described herein as compared to a control. In another embodiment, generating an immune response comprises an increase in target-specific CTL activity of about 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 15, 16, 17, 18, 19, 20, or more fold in a subject administered the adenovirus vectors as compared to a control.

[00361] Generating an immune response can comprise an increase in target antigen-specific HTL activity, such as proliferation of helper T-cells, of between 1.5 and 5 fold in a subject administered the adenovirus vectors that comprise nucleic acid encoding the target antigen as compared to an appropriate control. In another embodiment, generating an immune response comprises an increase in target-specific HTL activity of about 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 15, 16, 17, 18, 19, 20, or more fold as compared to a control. In this context, HTL activity may comprise an increase as described above, or decrease, in production of a particular cytokine, such as interferon-γ (IFN-γ), interleukin-1 (IL-1), IL-2, IL-3, IL-6, IL-7, IL-12, IL-15, tumor necrosis factor-a (TNF-a), granulocyte macrophage colony-stimulating factor (GM-CSF), granulocyte-colony stimulating factor (G-CSF), or other cytokine. In this regard, generating an immune response may comprise a shift from a Th2 type response to a Thl type response or in certain embodiments a shift from a Thl type response to a Th2 type response. In other embodiments, generating an immune response may comprise the stimulation of a predominantly Thl or a Th2 type response.

[00362] Generating an immune response can comprise an increase in target-specific antibody production of between 1.5 and 5 fold in a subject administered the adenovirus vectors as compared to an appropriate control. In another embodiment, generating an immune response comprises an increase in target-specific antibody production of about 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 15, 16, 17, 18, 19, 20, or more fold in a subject administered the adenovirus vector as compared to a control.

[00363] Thus, some embodiments provide methods for generating an immune response against a target antigen of interest comprising administering to the individual an adenovirus vector comprising: a) a replication defective adenovirus vector, wherein the adenovirus vector has a deletion in the E2b region, and b) a nucleic acid encoding the target antigen; and readministering the adenovirus vector at least once to the individual; thereby generating an immune response against the target antigen. In certain embodiments, the vector administered to the individual is not a gutted vector. In particular embodiments, the target antigen may be a wild-type protein, a fragment, a variant, or a variant fragment thereof. In some embodiments, the target antigen comprises MUC1, MUClc, MUCln, T, or CEA, a fragment, a variant, or a variant fragment thereof.

[00364] In a further embodiment, there is provided methods for generating an immune response against a target antigen in an individual, wherein the individual has preexisting immunity to Ad, by administering to the individual an adenovirus vector comprising: a) a replication defective adenovirus vector, wherein the adenovirus vector has a deletion in the E2b region, and b) a nucleic acid encoding the target antigen; and readministering the adenovirus vector at least once to the individual; thereby generating an immune response against the target antigen. In particular embodiments, the target antigen may be a wild-type protein, a fragment, a variant, or a variant fragment thereof. In some embodiments, the target antigen comprises MUC1, MUClc, MUCln, T, or CEA, a fragment, a variant, or a variant fragment thereof.

[00365] With regard to preexisting immunity to Ad, this can be determined using any suitable methods, such as antibody -based assays to test for the presence of Ad antibodies. Further, in certain embodiments, the methods include first determining that an individual has preexisting immunity to Ad then administering the E2b deleted adenovirus vectors as described herein.

[00366] One embodiment provides a method of generating an immune response against one or more target antigens in an individual comprising administering to the individual a first adenovirus vector comprising a replication defective adenovirus vector, wherein the adenovirus vector has a deletion in the E2b region, and a nucleic acid encoding at least one target antigen; administering to the individual a second adenovirus vector comprising a replication defective adenovirus vector, wherein the adenovirus vector has a deletion in the E2b region, and a nucleic acid encoding at least one target antigen, wherein the at least one target antigen of the second adenovirus vector is the same or different from the at least one target antigen of the first adenovirus vector. In particular embodiments, the target antigen may be a wild-type protein, a fragment, a variant, or a variant fragment thereof. In some embodiments, the target antigen comprises MUC1, MUClc, MUCln, T, or CEA, a fragment, a variant, or a variant fragment thereof.

[00367] Thus, multiple immunizations with the same E2b deleted adenovirus vector or multiple immunizations with different E2b deleted adenovirus vectors are contemplated in some

embodiments. In each case, the adenovirus vectors may comprise nucleic acid sequences that encode one or more target antigens as described elsewhere herein. In certain embodiments, the methods comprise multiple immunizations with an E2b deleted adenovirus encoding one target antigen, and re-administration of the same adenovirus vector multiple times, thereby inducing an immune response against the target antigen. In some embodiments, the target antigen comprises MUCl, MUClc, MUCln, T, or CEA, a fragment, a variant, or a variant fragment thereof.

[00368] In a further embodiment, the methods comprise immunization with a first adenovirus vector that encodes one or more target antigens, and then administration with a second adenovirus vector that encodes one or more target antigens that may be the same or different from those antigens encoded by the first adenovirus vector. In this regard, one of the encoded target antigens may be different or all of the encoded antigens may be different, or some may be the same and some may be different. Further, in certain embodiments, the methods include administering the first adenovirus vector multiple times and administering the second adenovirus multiple times. In this regard, the methods comprise administering the first adenovirus vector 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more times and administering the second adenovirus vector 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more times. The order of administration may comprise administering the first adenovirus one or multiple times in a row followed by administering the second adenovirus vector one or multiple times in a row. In certain embodiments, the methods include alternating administration of the first and the second adenovirus vectors as one administration each, two administrations each, three administrations each, and so on. In certain embodiments, the first and the second adenovirus vectors are administered simultaneously. In other embodiments, the first and the second adenovirus vectors are administered sequentially. In some embodiments, the target antigen comprises MUC1, MUClc, MUCln, T, or CEA, a fragment, a variant, or a variant fragment thereof.

[00369] As would be readily understood by the skilled artisan, more than two adenovirus vectors may be used in the methods. Three, 4, 5, 6, 7, 8, 9, 10 or more different adenovirus vectors may be used in the methods as described herein. In certain embodiments, the methods comprise

administering more than one E2b deleted adenovirus vector at a time. In this regard, immune responses against multiple target antigens of interest can be generated by administering multiple different adenovirus vectors simultaneously, each comprising nucleic acid sequences encoding one or more target antigens.

[00370] The adenovirus vectors can be used to generate an immune response against a cancer, such as carcinomas or sarcomas (e.g., solid tumors, lymphomas and leukemia). The adenovirus vectors can be used to generate an immune response against an infectious disease, such as an HPV infection, or a cancer, such as any CEA-expressing cancer, Brachyury-expressing cancer, MUCl-expessing cancer, HFV-associated cancer, an epithelial cancer, a neurologic cancer, melanoma, non-Hodgkin's lymphoma, Hodgkin's disease, leukemia, plasmocytomas, adenomas, gliomas, thymomas, breast cancer, prostate cancer, colorectal cancer, kidney cancer, renal cell carcinoma, uterine cancer, pancreatic cancer, esophageal cancer, lung cancer, ovarian cancer, cervical cancer, testicular cancer, gastric cancer, multiple myeloma, hepatoma, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL), gastrointestinal cancer, or other cancers.

[00371] Methods are also provided for treating or ameliorating the symptoms of any of the infectious diseases or cancers as described herein. The methods of treatment comprise administering the adenovirus vectors one or more times to individuals suffering from or at risk from suffering from an infectious disease or cancer as described herein. As such, some embodiments provide methods for vaccinating against infectious diseases or cancers in individuals who are at risk of developing such a disease. Individuals at risk may be individuals who may be exposed to an infectious agent at some time or have been previously exposed but do not yet have symptoms of infection or individuals having a genetic predisposition to developing a cancer or being particularly susceptible to an infectious agent. Individuals suffering from an infectious disease or cancer described herein may be determined to express and/or present a target antigen, which may be use to guide the therapies herein. For example, an example can be found to express and/or present a target antigen and an adenovirus vector encoding the target antigen, a variant, a fragment or a variant fragment thereof may be administered subsequently.

[00372] Some embodiments contemplate the use of adenovirus vectors for the in vivo delivery of nucleic acids encoding a target antigen, or a fragment, a variant, or a variant fragment thereof. Once injected into a subject, the nucleic acid sequence is expressed resulting in an immune response against the antigen encoded by the sequence. The adenovirus vector vaccine can be administered in an "effective amount", that is, an amount of adenovirus vector that is effective in a selected route or routes of administration to elicit an immune response as described elsewhere herein. An effective amount can induce an immune response effective to facilitate protection or treatment of the host against the target infectious agent or cancer. The amount of vector in each vaccine dose is selected as an amount which induces an immune, immunoprotective or other immunotherapeutic response without significant adverse effects generally associated with typical vaccines. Once vaccinated, subjects may be monitored to determine the efficacy of the vaccine treatment. Monitoring the efficacy of vaccination may be performed by any method known to a person of ordinary skill in the art. In some embodiments, blood or fluid samples may be assayed to detect levels of antibodies. In other embodiments, ELISpot assays may be performed to detect a cell -mediated immune response from circulating blood cells or from lymphoid tissue cells.

[00373] Routes and frequency of administration of the therapeutic compositions described herein, as well as dosage, may vary from individual to individual, and from disease to disease, and may be readily established using standard techniques. In general, the pharmaceutical compositions and vaccines may be administered by injection {e.g., intracutaneous, intramuscular, intravenous or subcutaneous), intranasally {e.g., by aspiration), in pill form {e.g., swallowing, suppository for vaginal or rectal delivery). In certain embodiments, between 1 and 10 doses may be administered over a 52 week period. In certain embodiments, 6 doses are administered, at intervals of 1 month, and further booster vaccinations may be given periodically thereafter. Alternate protocols may be appropriate for individual patients. As such, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more doses may be administered over a 1 year period or over shorter or longer periods, such as over 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 week periods. Doses may be administered at 1, 2, 3, 4, 5, or 6 week intervals or longer intervals.

[00374] A vaccine can be infused over a period of less than about 4 hours, and more preferably, over a period of less than about 3 hours. For example, the first 25-50 mg could be infused within 30 minutes, preferably even 15 min, and the remainder infused over the next 2-3 hrs. More generally, the dosage of an administered vaccine construct may be administered as one dosage every 2 or 3 weeks, repeated for a total of at least 3 dosages. Or, the construct may be administered twice per week for 4-6 weeks. The dosing schedule can optionally be repeated at other intervals and dosage may be given through various parenteral routes, with appropriate adjustment of the dose and schedule. Compositions can be administered to a patient in conjunction with (e.g., before, simultaneously, or following) any number of relevant treatment modalities.

[00375] A suitable dose is an amount of an adenovirus vector that, when administered as described above, is capable of promoting a target antigen immune response as described elsewhere herein. In certain embodiments, the immune response is at least 10-50% above the basal (i.e., untreated) level. In certain embodiments, the immune response is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 100, 110, 125, 150, 200, 250, 300, 400, 500 or more over the basal level. Such response can be monitored by measuring the target antigen(s) antibodies in a patient or by vaccine-dependent generation of cytolytic effector cells capable of killing patient tumor or infected cells in vitro, or other methods known in the art for monitoring immune responses. Such vaccines should also be capable of causing an immune response that leads to an improved clinical outcome of the disease in question in vaccinated patients as compared to non-vaccinated patients. In some embodiments, the improved clinical outcome comprises treating disease, reducing the symptoms of a disease, changing the progression of a disease, or extending life.

[00376] In general, an appropriate dosage and treatment regimen provides the adenovirus vectors in an amount sufficient to provide therapeutic and/or prophylactic benefit. Such a response can be monitored by establishing an improved clinical outcome for the particular disease being treated in treated patients as compared to non-treated patients. The monitoring data can be evaluated over time. The progression of a disease over time can be altered. Such improvements in clinical outcome would be readily recognized by a treating physician. Increases in preexisting immune responses to a target protein can generally correlate with an improved clinical outcome. Such immune responses may generally be evaluated using standard proliferation, cytotoxicity or cytokine assays, which may be performed using samples obtained from a patient before and after treatment.

[00377] While one advantage is the capability to administer multiple vaccinations with the same or different adenovirus vectors, particularly in individuals with preexisting immunity to Ad, the adenoviral vaccines may also be administered as part of a prime and boost regimen. A mixed modality priming and booster inoculation scheme may result in an enhanced immune response. Thus, one aspect is a method of priming a subject with a plasmid vaccine, such as a plasmid vector comprising a target antigen of interest, by administering the plasmid vaccine at least one time, allowing a predetermined length of time to pass, and then boosting by administering the adenovirus vector. Multiple primings, e.g., 1-4, may be employed, although more may be used. The length of time between priming and boost may typically vary from about four months to a year, but other time frames may be used. In certain embodiments, subjects may be primed 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times with plasmid vaccines, and then boosted 4 months later with the adenovirus vector. [00378] Any of the compositions provided herein may be administered to an individual.

"Individual" may be used interchangeably with "subject" or "patient." An individual may be a mammal, for example a human or animal such as a non-human primate, a rodent, a rabbit, a rat, a mouse, a horse, a donkey, a goat, a cat, a dog, a cow, a pig, or a sheep. In embodiments, the individual is a human. In embodiments, the individual is a fetus, an embryo, or a child. In some cases, the compositions provided herein are administered to a cell ex vivo. In some cases, the compositions provided herein are administered to an individual as a method of treating a disease or disorder. In some embodiments, the individual has a genetic disease. In some cases, the individual is at risk of having the disease, such as any of the diseases described herein. In some embodiments, the individual is at increased risk of having a disease or disorder caused by insufficient amount of a protein or insufficient activity of a protein. If an individual is "at an increased risk" of having a disease or disorder, the method involves preventative or prophylactic treatment. For example, an individual can be at an increased risk of having such a disease or disorder because of family history of the disease. Typically, individuals at an increased risk of having such a disease or disorder benefit from prophylactic treatment (e.g. , by preventing or delaying the onset or progression of the disease or disorder).

[00379] In some cases, a subject does not have a disease. In some cases, the treatment is administered before onset of a disease. A subject may have undetected disease. A subject may have a low disease burden. A subject may also have a high disease burden. In certain cases, a subject may be administered a treatment as described herein according to a grading scale. A grading scale can be a Gleason classification. A Gleason classification reflects how different tumor tissue is from normal prostate tissue. It uses a scale from 1 to 5. A physician gives a cancer a number based on the patterns and growth of the cancer cells. The lower the number, the more normal the cancer cells look and the lower the grade. The higher the number, the less normal the cancer cells look and the higher the grade. In certain cases, a treatment may be administered to a patient with a low Gleason score. Particularly, a patient with a Gleason score of 3 or below may be administered a treatment as described herein.

[00380] Various embodiments relate to compositions and methods for raising an immune response against one or more MUC1, MUClc, MUCln, T, or CEA antigens in selected patient populations. Accordingly, methods and compositions may target patients with a cancer including but not limited to carcinomas or sarcomas such as neurologic cancers, melanoma, non-Hodgkin's lymphoma, Hodgkin's disease, leukemia, plasmocytomas, adenomas, gliomas, thymomas, breast cancer, gastrointestinal cancer, prostate cancer, colorectal cancer, kidney cancer, renal cell carcinoma, uterine cancer, pancreatic cancer, esophageal cancer, lung cancer, ovarian cancer, cervical cancer, testicular cancer, gastric cancer, multiple myeloma, hepatoma, acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL), or other cancers can be targeted for therapy. In some cases, the targeted patient population may be limited to individuals having colorectal adenocarcinoma, metastatic colorectal cancer, advanced MUC1, MUClc, MUCln, T, or CEA expressing colorectal cancer, head and neck cancer, liver cancer, breast cancer, lung cancer, bladder cancer, or pancreas cancer. A histologically confirmed diagnosis of a selected cancer, for example colorectal adenocarcinoma, may be used. A particular disease stage or progression may be selected, for example, patients with one or more of a metastatic, recurrent, stage ΠΙ, or stage IV cancer may be selected for therapy with the methods and compositions. In some embodiments, patients may be required to have received and, optionally, progressed through other therapies including but not limited to fluoropyrimidine, irinotecan, oxaliplatin, bevacizumab, cetuximab, or panitumumab containing therapies. In some cases, individual's refusal to accept such therapies may allow the patient to be included in a therapy eligible pool with methods and compositions. In some embodiments, individuals to receive therapy using the methods and compositions may be required to have an estimated life expectancy of at least, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 18, 21, or 24 months. The patient pool to receive a therapy using the methods and compositions may be limited by age. For example, individuals who are older than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, 30, 35, 40, 50, 60, or more years old can be eligible for therapy with methods and compositions. For another example, individuals who are younger than 75, 70, 65, 60, 55, 50, 40, 35, 30, 25, 20, or fewer years old can be eligible for therapy with methods and compositions.

[00381] In some embodiments, patients receiving therapy using the methods and compositions are limited to individuals with adequate hematologic function, for example with one or more of a WBC count of at least 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000 or more per microliter, a hemoglobin level of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or higher g/dL, a platelet count of at least 50,000; 60,000; 70,000; 75,000; 90,000; 100,000; 110,000; 120,000; 130,000; 140,000; 150,000 or more per microliter; with a PT-INR value of less than or equal to 0.8, 1.0, 1.2, 1.3, 1.4, 1.5, 1.6, 1.8, 2.0, 2.5, 3.0, or higher, a PTT value of less than or equal to 1.2, 1.4, 1.5, 1.6, 1.8, 2.0 X ULN or more. In various embodiments, hematologic function indicator limits are chosen differently for individuals in different gender and age groups, for example 0-5, 5-10, 10-15, 15-18, 18-21, 21-30, 30-40, 40-50, 50-60, 60-70, 70-80 or older than 80.

[00382] In some embodiments, patients receiving therapy using the methods and compositions are limited to individuals with adequate renal and/or hepatic function, for example with one or more of a serum creatinine level of less than or equal to 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2 mg/dL or more, a bilirubin level of .8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2 mg/dL or more, while allowing a higher limit for Gilbert's syndrome, for example, less than or equal tol.5, 1.6, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, or 2.4 mg/dL, an ALT and AST value of less than or equal to less than or equal to 1.5, 2.0, 2.5, 3.0 x upper limit of normal (ULN) or more. In various embodiments, renal or hepatic function indicator limits are chosen differently for individuals in different gender and age groups, for example 0-5, 5-10, 10-15, 15-18, 18-21, 21-30, 30-40, 40-50, 50-60, 60-70, 70-80 or older than 80.

[00383] In some embodiments, the K-ras mutation status of individuals who are candidates for a therapy using the methods and compositions as described herein can be determined. Individuals with a preselected K-ras mutational status can be included in an eligible patient pool for therapies using the methods and compositions as described herein.

[00384] In various embodiments, patients receiving therapy using the methods and compositions as described herein are limited to individuals without concurrent cytotoxic chemotherapy or radiation therapy, a history of, or current, brain metastases, a history of autoimmune disease, such as but not restricted to, inflammatory bowel disease, systemic lupus erythematosus, ankylosing spondylitis, scleroderma, multiple sclerosis, thyroid disease and vitiligo, serious intercurrent chronic or acute illness, such as cardiac disease (NYHA class III or IV), or hepatic disease, a medical or

psychological impediment to probable compliance with the protocol, concurrent (or within the last 5 years) second malignancy other than non-melanoma skin cancer, cervical carcinoma in situ, controlled superficial bladder cancer, or other carcinoma in situ that has been treated, an active acute or chronic infection including: a urinary tract infection, HTV (e.g. as determined by ELISA and confirmed by Western Blot), and chronic hepatitis, or concurrent steroid therapy (or other immunosuppressives, such as azathioprine or cyclosporin A). In some cases, patients with at least 3, 4, 5, 6, 7, 8, 9, or 10 weeks of discontinuation of any steroid therapy (except that used as pre-medication for chemotherapy or contrast-enhanced studies) may be included in a pool of eligible individuals for therapy using the methods and compositions as described herein.

[00385] In some embodiments, patients receiving therapy using the methods and compositions as described herein include individuals with thyroid disease and vitiligo.

[00386] In various embodiments, samples, for example serum or urine samples, from the individuals or candidate individuals for a therapy using the methods and compositions as described herein may be collected. Samples may be collected before, during, and/or after the therapy for example, within 2, 4, 6, 8, 10 weeks prior to the start of the therapy, within 1 week, 10 day, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, or 12 weeks from the start of the therapy, within 2, 4, 6, 8, 10 weeks prior to the start of the therapy, within 1 week, 10 day, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 9 weeks, or 12 weeks from the start of the therapy, in 1 week, 10 day, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 9 weeks, or 12 weeks intervals during the therapy, in 1 month, 3 month, 6 month, 1 year, 2 year intervals after the therapy, within 1 month, 3 months, 6 months, 1 year, 2 years, or longer after the therapy, for a duration of 6 months, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years or longer. The samples may be tested for any of the hematologic, renal, or hepatic function indicators described herein as well as suitable others known in the art, for example a β-HCG for women with childbearing potential. In that regard, hematologic and biochemical tests, including cell blood counts with differential, PT, INR and PTT, tests measuring Na, K, CI, C0 2i BUN, creatinine, Ca, total protein, albumin, total bilirubin, alkaline phosphatase, AST, ALT and glucose may be used in some embodiments. In some embodiments, the presence or the amount of HTV antibody, Hepatitis BsAg, or Hepatitis C antibody are determined in a sample from individuals or candidate individuals for a therapy using the methods and compositions as described herein. Biological markers, such as antibodies to MUC1, MUClc, MUCln, T, or CEA or the neutralizing antibodies to Ad5 vector can be tested in a sample, such as serum, from individuals or candidate individuals for a therapy using the methods and compositions as described herein. In some cases, one or more samples, such as a blood sample can be collected and archived from an individuals or candidate individuals for a therapy using the methods and compositions as described herein. Collected samples can be assayed for immunologic evaluation. Individuals or candidate individuals for a therapy using the methods and compositions as described hereincan be evaluated in imaging studies, for example using CT scans or MRI of the chest, abdomen, or pelvis. Imaging studies can be performed before, during, or after therapy using the methods and compositions as described herein, during, and/or after the therapy, for example, within 2, 4, 6, 8, 10 weeks prior to the start of the therapy, within 1 week, 10 day, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, or 12 weeks from the start of the therapy, within 2, 4, 6, 8, 10 weeks prior to the start of the therapy, within 1 week, 10 day, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 9 weeks, or 12 weeks from the start of the therapy, in 1 week, 10 day, 2 week, 3 week, 4 week, 6 week, 8 week, 9 week, or 12 week intervals during the therapy, in 1 month, 3 month, 6 month, 1 year, 2 year intervals after the therapy, within 1 month, 3 months, 6 months, 1 year, 2 years, or longer after the therapy, for a duration of 6 months, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 years or longer.

Methods of Treating HPV-associated Diseases

[00387] In certain embodiments, there is provided a method of enhancing an immune response in an individual in need thereof, the method comprising: administering to the individual a first pharmaceutical composition comprising a replication-defective adenovirus vector comprising a nucleic acid sequence encoding an HPV antigen; and administering to the individual an immune checkpoint inhibitor. In certain embodiments, the method may be further defined as treating an HPV infection or an HPV-associated disease, such as an HPV-associated cancer, including, but not limited to, head and neck squamous cell carcinoma (HNSCC), oropharyngeal and tonsillar cancer, cervical cancer, or anal cancer.

[00388] Human papilloma virus (HPV)-associated HNSCC

[00389] Evidence has demonstrated that infection with high-risk HPV16 is associated with the development and progression of HPV-associated HNSCC and, more specifically, the HPV early

6 (E6) and early 7 (E7) genes contribute to cancer development. The prevalence of head and neck cancers in the United States is estimated to be about 370,000 and between 25% to 38% of these are HPV-associated HNSCC. Thus, the prevalence of HPV-associated HNSCC is estimated to range from 92,750 to 140,000 cases. A recent study on HPV-associated HNSCC estimated an incidence of about 35,000 new cases in the United States, with an expected 7,600 cancer related deaths annually despite current therapy. Thus, there remains an unmet medical need to investigate new treatment methods for this patient population. Based upon the estimated prevalence of HPV-associated HNSCC, this population qualifies for orphan product drug development by the FDA and Etubics has received orphan product designation for the development of a new immunotherapeutic vaccine (Ad5 [E1-, E2b-]-HPV-E6/E7) to treat HPV- associated HNSCC.

[00390] HIV and HPV-associated oropharyngeal and tonsillar cancer

[00391] Human papilloma virus (HPV) is responsible for as many as 100,000 cases of head and neck squamous cell carcinoma (HNSSC) worldwide per year. The majority of these are oropharyngeal and tonsillar cancers. In the United States, prevalence estimates of oropharynx HPV infection range from 9.2 to 18.6 percent. HPV typel6 (HPV16) is the most prevalent HPV found in oral carcinomas and is involved in the etiology of these cancers. The incidence of tonsillar cancer in the United States has increased by 2-3% per year from 1973 to 1995. HIV- infected individuals have a 2 to 6-fold increase in risk of developing oropharyngeal and tonsillar cancers. Despite significant advances in the therapy of AIDS, this pandemic continues to be responsible for devastating morbidity and mortality throughout the world, especially in regions with limited access to antiretroviral medications. HPV infection and disease has not dramatically declined since the introduction of potent combination therapy to control HIV and highly active antiretroviral therapy appears to have little beneficial effect on HPV-associated oral disease. Thus, it remains imperative to investigate new vaccines that can be applied to HIV and HIV- associated malignancies.

[00392] Certain aspects provide a therapeutic strategy for HIV-associated malignancy based on the pathogenic role of HPV. The vaccine to be used is based upon a new recombinant adenovirus serotype 5 (Ad5) vector platform (Ad5 [E1-, E2b-]) described below. This recombinant vector allows for the insertion of specific disease associated antigen genes that will be expressed after direct transfection of antigen presenting cells. Importantly, this new vaccine can be utilized in multiple homologous immunization regimens designed to stimulate potent cell mediated immune (CMI) responses against specific target antigens and has the potential to become an important immunotherapeutic agent in the battle against HIV/HPV-associated oropharyngeal and tonsillar malignancies. [00393] Patients with HPV associated HNSCC will be administered a multi-facetted treatment, and immunotherapy with the Etubics Ad5 [E1-, E2b-]-HPV-E6/E7 vaccine will play an important role in the armamentarium of treatments against this disease.

[00394] HPV associated Cervical Cancer

[00395] Cervical cancer is the second leading cause of cancer-related death in women. It is known that oncogenic human papillomavirus (HPV) plays a critical etiological role in anogenital cancers and at least 70% of cervical cancers are associated with type 16 (HPV- 16) or 18 (HPV- 18). HPV-16 and 18 are also the virus types with which the majority of vulval and vaginal precancer are associated. Vulvar intraepithelial neoplasia is a chronic premalignant disorder of the vulvar skin that is caused by high-risk types of human papillomavirus (HPV); HPV type 16 (HPV-16) is involved in more than 75% of cases. The lifetime risk of a woman acquiring any HPV infection is more than 80%. Half of women acquire cervical infection within 3 years of initiating sexual activity. About 90% of HPV infections are cleared by the immune system within 6-24 months. The prevalence of HPV infection in sexually active women is 10-20% and even higher in young women. HPV-16/18 bivalent (Cervarix) and HPV-6/11/16/18 quadrivalent (Gardasil) vaccines are highly effective in preventing vaccine-type HPV-related genital precancer in women who are HPV-negative at the time of vaccination. Although these vaccines are highly effective at preventing HPV infection, there is still a population of women who are not vaccinated and become HPV infected and thus are at high risk of developing neoplasia. In a recent meta-analysis study, there was no indication that the above HPV vaccines given to women with evidence of prior vaccine-type HPV exposure can prevent premalignant lesions related to these HPV types over a 3 to 4-year time frame. It is this population of women that are believed to benefit from vaccination with this new adenoviral vaccine (Ad5 [E1-, E2b-]-HPV- E6/E7 vaccine) designed to prevent development of HPV associated cancer.

Dosages and Administration

[00396] Compositions and methods as described hereincontemplate various dosage and

administration regimens during therapy. Patients may receive one or more replication defective adenovirus or adenovirus vector, for example Ad5 [E1-, E2B-]-CEA(6D), Ad5 [E1-, E2b-]-MUCl, Ad5 [E1-, E2b-]-MUClc, Ad5 [E1-, E2b-]-MUCln, Ad5 [E1-, E2b-]-T (i.e., Ad5 [E1-, E2b-]- Brachyury) that is capable of raising an immune response in an individual against a target antigen described herein. In various embodiments, the replication defective adenovirus is administered at a dose that suitable for effecting such immune response. In some cases, the replication defective adenovirus is administered at a dose that is greater than or equal to lxl 0 9 , 2 xlO 9 , 3 xlO 9 , 4 xlO 9 , 5 xlO 9 , 6 xlO 9 , 7 xlO 9 , 8 xlO 9 , 9 xlO 9 , lxlO 10 , 2 xlO 10 , 3 xlO 10 , 4 xlO 10 , 5 xlO 10 , 6 xlO 10 , 7 xlO 10 , 8 xlO 10 , 9 xlO 10 , 1 xlO 11 , 2 xlO 11 , 3 xlO 11 , 4 xlO 11 , 5xl0 n , 6 xlO 11 , 7 xlO 11 , 8 xlO 11 , 9 xlO 11 , lxlO 12 , 1.5 xlO 12 , 2 xlO 12 , 3 xlO 12 , or more virus particles (VP) per immunization. In some cases, the replication defective adenovirus is administered at a dose that is less than or equal to lxl 0 9 , 2 xlO 9 , 3 xlO 9 , 4 xlO 9 , 5 xlO 9 , 6 xlO 9 , 7 xlO 9 , 8 xlO 9 , 9 xlO 9 , lxlO 10 , 2 xlO 10 , 3 xlO 10 , 4 xlO 10 , 5 xlO 10 , 6 xlO 10 , 7 xlO 10 , 8 xlO 10 , 9 xlO 10 , 1 xlO 11 , 2 xlO 11 , 3 xlO 11 , 4 xlO 11 , 5xlO n , 6 xlO 11 , 7 xlO 11 , 8 xlO 11 , 9 xlO , 1x10 , 1.5 xlO , 2 xlO , 3 xlO , or more virus particles per immunization. In various embodiments, a desired dose described herein is administered in a suitable volume of formulation buffer, for example a volume of about 0.1-10 mL, 0.2-8mL, 0.3-7mL, 0.4-6 mL, 0.5-5 mL, 0.6-4 mL, 0.7-3 mL, 0.8-2 mL, 0.9-1.5 mL, 0.95-1.2 mL, or 1.0-1.1 mL. Those of skill in the art appreciate that the volume may fall within any range bounded by any of these values (e.g., about 0.5 mL to about 1.1 mL). Administration of virus particles can be through a variety of suitable paths for delivery, for example it can be by injection (e.g., intradermally, intracutaneously, intramuscularly, intravenously or subcutaneously), intranasally (e.g., by aspiration), in pill form (e.g. swallowing, suppository for vaginal or rectal delivery. In some embodiments, a subcutaneous delivery may be preferred and can offer greater access to dendritic cells.

[00397] Administration of virus particles to an individual may be repeated. Repeated deliveries of virus particles may follow a schedule or alternatively, may be performed on an as needed basis. For example, an individual's immunity against a target antigen, for example MUCl, T and/or CEA, may be tested and replenished as necessary with additional deliveries. In some embodiments, schedules for delivery include administrations of virus particles at regular intervals. Joint delivery regimens may be designed comprising one or more of a period with a schedule and/or a period of need based administration assessed prior to administration. For example, a therapy regimen may include an administration, such as subcutaneous administration once every three weeks then another immunotherapy treatment every three months until removed from therapy for any reason including death. Another example regimen comprises three administrations every three weeks then another set of three immunotherapy treatments every three months. Another example regimen comprises a first period with a first number of administrations at a first frequency, a second period with a second number of administrations at a second frequency, a third period with a third number of

administrations at a third frequency, etc., and optionally one or more periods with undetermined number of administrations on an as needed basis. The number of administrations in each period can be independently selected and can for example be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more. The frequency of the administration in each period can also be independently selected, can for example be about every day, every other day, every third day, twice a week, once a week, once every other week, every three weeks, every month, every six weeks, every other month, every third month, every fourth month, every fifth month, every sixth month, once a year etc. The therapy can take a total period of up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 30, 36 months or more. The scheduled interval between immunizations may be modified so that the interval between immunizations is revised by up to a fifth, a fourth, a third, or half of the interval. For example, for a 3 -week interval schedule, an immunization may be repeated between 20 and 28 days (3 weeks -1 day to 3 weeks +7 days). For the first 3 immunizations, if the second and/or third immunization is delayed, the subsequent immunizations may be shifted allowing a minimum amount of buffer between immunizations. For example, for a three week interval schedule, if an immunization is delayed, the subsequent immunization may be scheduled to occur no earlier than 17, 18, 19, or 20 days after the previous immunization.

[00398] Compositions, such as Ad Ad5 [E1-, E2B-]-CEA(6D), Ad5 [E1-, E2B-]-MUCl, Ad5 [E1-, E2B-]-MUClc, Ad5 [E1-, E2B-]-MUCln, Ad5 [E1-, E2B-]-T virus particles, can be provided in various states, for example, at room temperature, on ice, or frozen. Compositions may be provided in a container of a suitable size, for example a vial of 2 mL vial. In one embodiment, a 2-ml vial with 1.0 mL of extractable vaccine contains 5xl0 n total virus particles/mL. Storage conditions including temperature and humidity may vary. For example, compositions for use in therapy may be stored at room temperature, 4 °C, -20 °C, or lower.

[00399] In various embodiments, general evaluations are performed on the individuals receiving treatment according to the methods and compositions as described herein. One or more of any tests may be performed as needed or in a scheduled basis, such as on weeks 0, 3, 6 etc. A different set of tests may be performed concurrent with immunization vs. at time points without immunization.

[00400] General evaluations may include one or more of medical history, ECOG Performance Score, Karnofsky performance status, and complete physical examination with weight by the attending physician. Any other treatments, medications, biologies, or blood products that the patient is receiving or has received since the last visit may be recorded. Patients may be followed at the clinic for a suitable period, for example approximately 30 minutes, following receipt of vaccine to monitor for any adverse reactions. Local and systemic reactogenicity after each dose of vaccine will may be assessed daily for a selected time, for example for 3 days (on the day of immunization and 2 days thereafter). Diary cards may be used to report symptoms and a ruler may be used to measure local reactogenicity. Immunization injection sites may be assessed. CT scans or MRI of the chest, abdomen, and pelvis may be performed.

[00401] In various embodiments, hematological and biochemical evaluations are performed on the individuals receiving treatment according to the methods and compositions as described herein. One or more of any tests may be performed as needed or in a scheduled basis, such as on weeks 0, 3, 6 etc. A different set of tests may be performed concurrent with immunization vs. at time points without immunization. Hematological and biochemical evaluations may include one or more of blood test for chemistry and hematology, CBC with differential, Na, K, CI, C0 2i BUN, creatinine, Ca, total protein, albumin, total bilirubin, alkaline phosphatase, AST, ALT, glucose, and ANA [00402] In various embodiments, biological markers are evaluated on individuals receiving treatment according to the methods and compositions as described herein. One or more of any tests may be performed as needed or in a scheduled basis, such as on weeks 0, 3, 6 etc. A different set of tests may be performed concurrent with immunization vs. at time points without immunization.

[00403] Biological marker evaluations may include one or more of measuring antibodies to MUCl, MUClc, MUCln, CEA or the Ad5 vector, from a serum sample of adequate volume, for example about 5ml Biomarkers (e.g., CEA or CA15-3) may be reviewed if determined and available.

[00404] In various embodiments, an immunological assessment is performed on individuals receiving treatment according to the methods and compositions as described herein. One or more of any tests may be performed as needed or in a scheduled basis, such as on weeks 0, 3, 6 etc. A different set of tests may be performed concurrent with immunization vs. at time points without immunization.

[00405] Peripheral blood, for example about 90mL may be drawn prior to each immunization and at a time after at least some of the immunizations, to determine whether there is an effect on the immune response at specific time points during the study and/or after a specific number of immunizations. Immunological assessment may include one or more of assaying peripheral blood mononuclear cells (PBMC) for T-cell responses to MUCl, MUClc, MUCln, T or CEA using ELISpot, proliferation assays, multi-parameter flow cytometric analysis, and cytoxicity assays. Serum from each blood draw may be archived and sent and determined.

[00406] In various embodiments, a tumor assessment is performed on individuals receiving treatment according to the methods and compositions as described herein. One or more of any tests may be performed as needed or in a scheduled basis, such as prior to treatment, on weeks 0, 3, 6 etc. A different set of tests may be performed concurrent with immunization vs. at time points without immunization. Tumor assessment may include one or more of CT or MRI scans of chest, abdomen, or pelvis performed prior to treatment, at a time after at least some of the immunizations and at approximately every three months following the completion of a selected number, for example 2, 3, or 4, of first treatments and for example until removal from treatment.

[00407] Immune responses against a target antigen described herein, such as CEA, may be evaluated from a sample, such as a peripheral blood sample of an individual using one or more suitable tests for immune response, such as ELISpot, cytokine flow cytometry, or antibody response. A positive immune response can be determined by measuring a T-cell response. A T-cell response can be considered positive if the mean number of spots adjusted for background in six wells with antigen exceeds the number of spots in six control wells by 10 and the difference between single values of the six wells containing antigen and the six control wells is statistically significant at a level of p<0.05 using the Student's t-test. Immunogenicity assays may occur prior to each immunization and at scheduled time points during the period of the treatment. For example, a time point for an immunogenicity assay at around week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 20 , 24, 30, 36, or 48 of a treatment may be scheduled even without a scheduled immunization at this time. In some cases, an individual may be considered evaluable for immune response if they receive at least a minimum number of immunizations, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, or more immunizations.

[00408] In some embodiments, disease progression or clinical response determination is made according to the RECIST 1.1 criteria among patients with measurable/evaluable disease. In some embodiments, therapies using the methods and compositions as described herein affect a Complete Response (CR; disappearance of all target lesions for target lesions or disappearance of all non-target lesions and normalization of tumor marker level for non-target lesions) in an individual receiving the therapy. In some embodiments, therapies using the methods and compositions affect a Partial Response (PR; at least a 30% decrease in the sum of the LD of target lesions, taking as reference the baseline sum LD for target lesions) in an individual receiving the therapy.

[00409] In some embodiments, therapies using the methods and compositions affect a Stable Disease (SD; neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum LD since the treatment started for target lesions) in an individual receiving the therapy. In some embodiments, therapies using the methods and

compositions as described herein affect an Incomplete Response/ Stable Disease (SD; persistence of one or more non-target lesion(s) or/and maintenance of tumor marker level above the normal limits for non-target lesions) in an individual receiving the therapy. In some embodiments, therapies using the methods and compositions as described herein affect a Progressive Disease (PD; at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started or the appearance of one or more new lesions for target lesions or persistence of one or more non-target lesion(s) or/and maintenance of tumor marker level above the normal limits for non-target lesions) in an individual receiving the therapy.

Additional Compositions and Methods

[00410] Some embodiments provide combination multi -targeted vaccines, immunotherapies and methods for enhanced therapeutic response to complex diseases such as infectious diseases and cancers. Certain embodiments provide compositions, methods and kits for generating an immune response in an individual to fight infectious diseases and cancer. Certain embodiments provide compositions, methods and kits for generating an immune response against a target antigen or cells expressing or presenting a target antigen or a target antigen signature comprising at least one target antigen.

[00411] It has been discovered that Ad5 [E1 -, E2b-] vectors are not only are safer than, but appear to be superior to Ad5 [E1-] vectors in regard to induction of antigen specific immune responses, making them much better suitable as a platform to deliver MUC1, T and/or CEA vaccines that can result in a clinical response. In other cases, immune induction may take months. Ad5 [E1 -, E2b-] vectors not only are safer than, but appear to be superior to Ad5 [E1-] vectors in regard to induction of antigen specific immune responses, making them much better suitable as a platform to deliver MUClc, T and/or CEA vaccines that can result in a clinical response.

[00412] Certain embodiments use the new Ad5 [E1-, E2b-] vector system to deliver a long sought- after need for a develop a therapeutic vaccine against MUC1, T and/or CEA, overcome barriers found with other Ad5 systems and permit the immunization of people who have previously been exposed to Ad5.

[00413] To address the low immunogenicity of self-tumor antigens, a variety of advanced, multi- component vaccination strategies including co-administration of adjuvants and immune stimulating cytokines are provided. Some embodiments relate to recombinant viral vectors that provide innate pro-inflammatory signals, while simultaneously engineered to express the antigen of interest. Of particular interest are adenovirus serotype-5 (Ad5)-based immunotherapeutics that have been repeatedly used in humans to induce robust T-cell-mediated immune (CMI) responses, all while maintaining an extensive safety profile.

[00414] In one aspect, a composition is provided comprising a recombinant replication defective viral vector comprising a sequence encoding a MUCl-C antigen, wherein the sequence encoding the MUCl-C antigen has at least 80% sequence identity to SEQ ID NO:5 or SEQ ID NO:6. In some embodiments, the MUCl-C antigen comprises a sequence with at least 80% sequence identity to SEQ ID NO:9.

[00415] In one aspect, a composition is provided comprising a recombinant replication defective viral vector comprising a sequence encoding a Brachyury antigen, wherein sequence encoding the Brachyury antigen has at least 80% sequence identity to SEQ ID NO: 7 or SEQ ID NO: 8. In some embodiments, an immune response against the antigen or cells expressing the antigen is induced in a human administered the viral vector.

[00416] In one aspect, a composition is provided comprising a recombinant replication defective viral vector comprising a sequence encoding at least two antigens selected from the group consisting of a MUCl-C antigen, a Brachyury antigen, and a CEA antigen. In some embodiments, an immune response against the at least two antigens or cells expressing the at least two antigens is induced in a human administered the viral vector.

[00417] In some embodiments, the immune response comprises generation of an antibody to the antigen. In some embodiments, the immune response comprises cell mediated immunity (CMI). In some embodiments, the sequence encoding the MUCl-C antigen has at least 80% sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6. In some embodiments, the sequence encoding the Brachyury antigen has at least 80% sequence identity to SEQ ID NO: 7 or SEQ ID NO: 8. In some embodiments, the sequence encoding the CEA antigen has at least 80% sequence identity to SEQ ID NO: 1, SEQ ID NO:2, or SEQ ID NO: 3. In some embodiments, the antigen comprises a modification of 25, 15, 10, 5, or less amino acids. In some embodiments, the antigen comprises a modification in 1 amino acid. In some embodiments, the recombinant viral vector is selected from the group consisting of: retrovirus, lentivirus, cytomegalovirus, Sendai virus, HPV virus, and adenovirus. In some embodiments, the recombinant viral vector comprises a replication defective adenovirus vector. In some embodiments, the recombinant viral vector comprises a replication defective adenovirus 5 vector. In some embodiments, the replication defective adenovirus vector comprises a deletion in an E2b gene region. In some embodiments, the replication defective adenovirus vector comprises a deletion in an El gene region. In some embodiments, the replication defective adenovirus vector comprises a deletion in an E3 gene region. In some embodiments, the replication defective adenovirus vector comprises a deletion in an E4 gene region. In some embodiments, the recombinant viral vector effects overexpression of the antigen in transfected cells. In some embodiments, the recombinant viral induces a specific immune response against cells expressing the antigen in a human that is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 fold over basal. In some embodiments, the human has an inverse Ad5 neutralizing antibody titer of greater than 50, 75, 100, 125, 150, 160, 175, or 200. In some embodiments, the human has an inverse Ad5 neutralizing antibody titer of greater than 250, 500, 750, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, or 4767. In some embodiments, the immune response is measured as antigen specific antibody response.

[00418] In some embodiments, the immune response is measured as antigen specific cell-mediated immunity (CMI). In some embodiments, the immune response is measured as antigen specific IFN-γ secretion. In some embodiments, the immune response is measured as antigen specific IL-2 secretion. In some embodiments, the immune response against the antigen is measured by ELISpot assay. In some embodiments, the antigen specific CMI is greater than25, 50, 75, 100, 150, 200, 250, or 300 IFN-γ spot forming cells (SFC) per 10 6 peripheral blood mononuclear cells (PBMC). In some embodiments, the immune response is measured by T-cell lysis of CAP-1 pulsed antigen-presenting cells, allogeneic antigen expressing cells from a tumor cell line or from an autologous tumor. In some embodiments, the composition further comprises an immunogenic component. In some embodiments, the immunogenic component comprises a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13. In some embodiments, the immunogenic component is selected from the group consisting of IL-7, a nucleic acid encoding IL-7, a protein with substantial identity to IL-7, and a nucleic acid encoding a protein with substantial identity to IL-7. In some embodiments, the CEA antigen comprises a modification that comprises a substitution of aspartate at a position corresponding to position 610 in SEQ ID NO: 3. In some embodiments, the composition further comprises a molecular composition comprising an immune pathway checkpoint modulator. In some embodiments, the immune pathway checkpoint modulator activates or potentiates an immune response. In some embodiments, the immune pathway checkpoint modulator inhibits an immune response inhibitor. In some embodiments, the immune pathway checkpoint inhibits an immune response. In some embodiments, the immune pathway checkpoint modulator targets an endogenous immune pathway checkpoint protein or fragment thereof selected from the group consisting of: PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TIM3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, and CD244. In some embodiments, the immune pathway checkpoint modulator targets a PD1 protein. In some embodiments, the molecular composition comprises siRNAs, antisense, small molecules, mimic, a recombinant form of a ligand, a recombinant form of a receptor, antibodies, or a combination thereof.

[00419] In one aspect, a method of selecting a human for administration of the compositions is provided comprising: determining a HLA subtype of the human; and administering the composition to the human, if the HLA subtype is determined to be one of a preselected subgroup of HLA subtypes. In some embodiments, the preselected subgroup of HLA subtypes comprises one or more of HLA-A2, HLA- A3, and HLA-A24.

[00420] In one aspect, a method of treating a human for cancer or an infectious disease is provided comprising administering the recombinant viral vector to the human.

[00421] In one aspect, a method of generating an immune response in a human to MUC1 -C, Brachyury, CEA, or any combination thereof is provided comprising administering to the human the composition. In some embodiments, the administering step is repeated at least once. In some embodiments, the administering step is repeated after about 2, 3, 4, 5, or 6 weeks following a previous administering step. In some embodiments, the administering step is repeated after about 2, 3, 4, 5, or 6 months following a previous administering step. In some embodiments, the

administering step is repeated twice.

[00422] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human a total of 3 times, in about 3 week intervals, a first composition comprising a first replication defective adenovirus vector encoding a MUC1-C antigen; and during the second phase, administering to the human a total of 3 times, in about 3 month intervals, a second composition comprising a second replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing the MUC1-C antigen.

[00423] In one aspect, a method of treatment is provided comprising: selecting a first phase and a second phase of treatment; during the first phase, administering to a human a total of 3 times, in about 3 week intervals, a first composition comprising a first replication defective adenovirus vector encoding a Brachyury antigen; and during the second phase, administering to the human a total of 3 times, in about 3 month intervals, a second composition comprising a second replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing the Brachyury antigen.

[00424] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human a total of 3 times, in about 3 week intervals, a first composition comprising a first replication defective adenovirus vector encoding at least two antigens selected from the group consisting of a MUC1-C antigen, a Brachyury antigen, and a CEA antigen; and during the second phase, administering to the human a total of 3 times, in about 3 month intervals, a second composition comprising a second replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing the at least two antigens. In some embodiments, the second phase starts about 3 months after the end of the first phase.

[00425] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human, a total of n times, a first composition comprising a first replication defective adenovirus vector encoding a Brachyury antigen; during the second phase, administering the human, a total of m times, a second composition comprising a second replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing the Brachyury antigen.

[00426] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human, a total of n times, a first composition comprising a first replication defective adenovirus vector encoding a MUC1-C antigen; during the second phase, administering the human, a total of m times, a second composition comprising a second replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing the MUC1-C antigen.

[00427] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human, a total of n times, a first composition comprising a first replication defective adenovirus vector encoding at least two antigens selected from the group consisting of a MUC1 -C antigen, a Brachyury antigen, and a CEA antigen; during the second phase, administering the human, a total of m times, a second composition comprising a second replication defective adenovirus vector encoding the at least two antigens that induces an immune response in a human against cells expressing the at least two antigens. In some embodiments, n is greater than 1. In some embodiments, n is 3. In some embodiments, m is greater than 1. In some embodiments, m is 3. In some embodiments, the first phase is at least 2, 3, 4, 5, 6, 7, or 8 weeks. In some embodiments, the second phase is at least 2, 3, 4, 5, 6, 7, or 8 months. In some embodiments, the second phase starts 3-16 weeks after first phase ends. In some embodiments, in the first phase two administrations of the replication defective adenovirus are at least 18 days apart. In some embodiments, in the first phase two administrations of the replication defective adenovirus are about 21 days apart. In some embodiments, in the first phase two administrations of the replication defective adenovirus are at most 24 days apart. In some embodiments, in the second phase two administrations of the replication defective adenovirus are at least 10 weeks apart. In some embodiments, in the second phase two administrations of the replication defective adenovirus are about 13 weeks apart. In some embodiments, in the second phase two administrations of the replication defective adenovirus are at most 16 weeks apart. In some embodiments, the method further comprises administering a molecular composition comprising an immune pathway checkpoint modulator.

[00428] In one aspect, a method of treatment is provided comprising: selecting a first phase of treatment and a second phase of treatment; during the first phase, administering to a human, a total of n times, a first composition comprising a first replication defective adenovirus vector encoding an antigen that induces an immune response in a human against cells expressing a MUC1 -C,

Brachyury, or CEA antigen; and during the second phase, administering the human, a total of m times, a second composition comprising a second replication defective adenovirus vector encoding an antigen that is capable of inducing an immune response directed towards cells expressing MUC1 - C, Brachyury, or CEA antigen in a human; wherein a molecular composition comprising and an immune pathway checkpoint modulator is administered during the first phase, the second phase, or both.

[00429] In one aspect, a method of treating a subject in need thereof is provided, comprising administering to the subject: (a) a recombinant replication deficient adenovirus vector encoding (i) a MUC1-C antigen, (ii) a Brachyury antigen, or (iii) at least two antigens selected from the group consisting of a MUC1-C antigen, a Brachyury antigen, and a CEA antigen; and (b) a molecular composition comprising an immune pathway checkpoint modulator; thereby generating an immune response in the subject. In some embodiments, (a) and (b) are administered in series. In some embodiments, (a) and (b) are administered at the same time. In some embodiments, (a) and (b) are administered a month apart.

[00430] In some embodiments, the immune pathway checkpoint modulator activates or potentiates an immune response. In some embodiments, the immune pathway checkpoint modulator inhibits an immune response inhibitor. In some embodiments, the immune pathway checkpoint inhibits an immune response. In some embodiments, the immune pathway checkpoint modulator targets an endogenous immune pathway checkpoint protein or fragment thereof selected from the group consisting of: PDl, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7- H4, BTLA, HVEM, KIR, TCR, LAG3, CD 137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TEVI3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, and CD244. In some embodiments, the immune pathway checkpoint modulator targets a PDl protein. In some embodiments, the molecular composition comprises siRNAs, antisense, small molecules, mimic, a recombinant form of a ligand, a recombinant form of a receptor, antibodies, or a combination thereof.

[00431] In some embodiments, the immune response is increased at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 fold. In some embodiments, the first replication defective adenovirus vector and the second replication defective adenovirus vector are the same. In some embodiments, the first replication defective adenovirus vector comprises a replication defective adenovirus 5 vector. In some embodiments, the second replication defective adenovirus vector comprises a replication defective adenovirus 5 vector. In some embodiments, the human expresses a human leukocyte antigen of serotype HLA-A2, HLA-A3, or HLA-A24. In some embodiments, the first replication defective adenovirus vector comprises a deletion in an E2b gene region. In some embodiments, the first replication defective adenovirus vector further comprises a deletion in an El gene region. In some embodiments, the first replication defective adenovirus vector further comprises a deletion in an E3 gene region. In some embodiments, the first replication defective adenovirus vector further comprises a deletion in an E4 gene region. In some embodiments, the second replication defective adenovirus vector comprises a deletion in an E2b gene region. In some embodiments, the second replication defective adenovirus vector further comprises a deletion in an El gene region. In some embodiments, the second replication defective adenovirus vector further comprises a deletion in an E3 gene region. In some embodiments, the second replication defective adenovirus vector further comprises a deletion in an E4 gene region. In some embodiments, the first composition, the second composition, or both, comprises at least l .OxlO 11 , 2.0xlO n , 3.0xl0 n , 3.5xl0 n , 4.0xlO n , 4.5xlO n , 4.8xlO n , 4.9xlO n , 4.95xlO n , or 4.99xlO n virus particles comprising the recombinant nucleic acid vector. In some embodiments, the first composition, the second composition, or both, comprises at most 7.0xlO n , 6.5xlO n , 6.0xlO n , 5.5xl0 n , 5.2xlO n , 5.1xl0 n , 5.05xl0 n , or 5.01xl0 n virus particles. In some embodiments, the first composition, the second composition, or both, comprises 1.0-7.0xlO n or 1.0-5.5xl0 n virus particles. In some embodiments, the first composition, the second composition, or both, comprises 4.5 -5.5x10 11 virus particles. In some embodiments, the first composition, the second composition, or both, comprises 4.8-5.2xlO n virus particles. In some embodiments, the first composition, the second composition, or both, comprises 4.9-5. lxlO 11 virus particles. In some embodiments, the first composition, the second composition, or both, comprises 4.95-5.05xlO n virus particles. In some embodiments, the first composition, the second composition, or both, comprises 4.99-5.01 x 10 11 virus particles. In some embodiments, the first phase is at least 2, 3, 4, 5, 6, 7, or 8 weeks. In some embodiments, the second phase is at least 2, 3, 4, 5, 6, 7, or 8 months. In some embodiments, the immune response is measured as antigen specific antibody response. In some embodiments, the immune response is measured as antigen specific cell-mediated immunity (CMI). In some embodiments, the immune response is measured as antigen specific IFN-γ secretion. In some embodiments, the immune response is measured as antigen specific IL-2 secretion. In some embodiments, the immune response against the antigen is measured by ELISpot assay. In some embodiments, the antigen specific CMI is greater than 25, 50, 75, 100, 150, 200, 250, or 300 IFN-γ spot forming cells (SFC) per 10 6 peripheral blood mononuclear cells (PBMC). In some embodiments, the immune response is measured by T-cell lysis of CAP-1 pulsed antigen-presenting cells, allogeneic antigen expressing cells from a tumor cell line or from an autologous tumor. In some embodiments, a first or a second replication defective adenovirus infects dendritic cells in the human and wherein the infected dendritic cells present the antigen, thereby inducing the immune response. In some embodiments, the administering steps comprise subcutaneous (sc) administration. In some embodiments, the human carries an inverse Ad5 neutralizing antibody titer that is of greater than 50, 75, 100, 125, 150, 160, 175, 200, 225, 250, 275, or 300 prior to the administering step. In some embodiments, the human has an inverse Ad5 neutralizing antibody titer of greater than 250, 500, 750, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, or 4767. In some embodiments, the human is not concurrently being treated by any one of steroids, corticosteroids, and

immunosuppressive agents. In some embodiments, the human does not have an autoimmune disease. In some embodiments, the human does not have inflammatory bowel disease, systemic lupus erythematosus, ankylosing spondylitis, scleroderma, multiple sclerosis, viral hepatitis, or HIV. In some embodiments, the human has or may have in the future an infectious disease. In some embodiments, the human has autoimmune related thyroid disease or vitiligo. In some embodiments, the human has or may have in the future a proliferative disease cancer. In some embodiments, the human has colorectal adenocarcinoma, metastatic colorectal cancer, advanced MUC1-C, Brachyury, or CEA expressing colorectal cancer, breast cancer, lung cancer, bladder cancer, or pancreas cancer. In some embodiments, the human has at least 1, 2, or 3 sites of metastatic disease. In some embodiments, the human comprises cells overexpressing MUC1-C, Brachyury, or CEA. In some embodiments, the cells overexpressing MUC1-C, Brachyury, or CEA, overexpress the MUC1-C, Brachyury, or CEA by at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 times over a baseline MUC1-C, Brachyury, or CEA expression in a non-cancer cell. In some embodiments, the cells overexpressing MUC1-C, Brachyury, or CEA comprise cancer cells. In some embodiments, the subject has a diagnosed disease predisposition. In some embodiments, the subject has a stable disease. In some

embodiments, the subject has a genetic predisposition for a disease. In some embodiments, the disease is a cancer. In some embodiments, the cancer is selected from the group consisting of prostate cancer, colon cancer, breast cancer, or gastric cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the subject has a Gleason score of 6 or less. In some embodiments, the subject has a Gleason score greater than 6. In some embodiments, the first or the second replication defective adenovirus vector comprises a sequence with at least 80% sequence identity to SEQ ID NO: 3. In some embodiments, the first or the second replication defective adenovirus vector comprises a region with at least 80% sequence identity to a region in SEQ ID NO:3 selected from 26048-26177, 26063-26141, 1-103, 54-103, 32214-32315, and 32214-32262. In some embodiments, the first or the second replication defective adenovirus vector comprises a region with at least 80% sequence identity to a region in SEQ ID NO:3 between positions 1057 and 3165. In some embodiments, the first or second replication defective adenovirus vector comprises a sequence encoding a MUC1-C, Brachyury, or CEA antigen; wherein the MUC1-C antigen is encoded by a sequence with at least 80% sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6; wherein the Brachyury antigen is encoded by a sequence with at least 80% sequence identity to SEQ ID NO:7 or SEQ ID NO: 8; wherein the CEA antigen is encoded by a sequence with at least 80% sequence identity to SEQ ID NO: l, SEQ ID NO:2, or SEQ ID NO:3.

[00432] In one aspect a kit for inducing an immune response in a human is provided comprising: a composition comprising a therapeutic solution of a volume in the range of 0.8-1.2 mL, the therapeutic solution comprising at least l.OxlO 11 virus particles; wherein the virus particles comprise a recombinant replication defective adenovirus vector; a composition comprising of a therapeutic solution of a molecular composition comprising an immune pathway checkpoint modulator and; instructions.

[00433] In some embodiments, the therapeutic solution comprises 1.0-5.5x10 11 virus particles. In some embodiments, adenovirus vector is capable of effecting overexpression of the modified MUC1-C, Brachyury, or CEA in transfected cells. In some embodiments, therapeutic solution comprises a first, second and third replication defective adenovirus vector each comprising an antigen selected from the group consisting of a MUC1-C antigen, a Brachyury antigen, a CEA antigen, and combinations thereof. In some embodiments, the adenovirus vector comprises a nucleic acid sequence encoding an antigen that induces a specific immune response against MUC1 -C, Brachyury, or CEA expressing cells in a human. In some embodiments, the immune pathway checkpoint modulator targets an endogenous immune pathway checkpoint protein or fragment thereof selected from the group consisting of: PD1, PDL1, PDL2, CD28, CD80, CD86, CTLA4, B7RP1, ICOS, B7RPI, B7-H3, B7-H4, BTLA, HVEM, KIR, TCR, LAG3, CD137, CD137L, OX40, OX40L, CD27, CD70, CD40, CD40L, TEVI3, GAL9, ADORA, CD276, VTCN1, IDOl, KIR3DL1, HAVCR2, VISTA, and CD244. In some embodiments, the molecular composition comprises siRNAs, antisense, small molecules, mimic, a recombinant form of a ligand, a recombinant form of a receptor, antibodies, or a combination thereof. In some embodiments, the instructions are for the treatment of a proliferative disease or cancer. In some embodiments, the instructions are for the treatment of an infectious disease. In some embodiments, the adenovirus vector comprises a replication defective adenovirus 5 vector. In some embodiments, the therapeutic solution comprises at least l.OxlO 11 , 2.0xlO n , 3.0xl0 n , 3.5xl0 n , 4.0xlO n , 4.5xlO n , 4.8xlO n , 4.9xlO n , 4.95xlO n , or 4.99xlO n virus particles comprising the recombinant nucleic acid vector. In some embodiments, the therapeutic solution comprises at most 7.0xlO n , 6.5xlO n , 6.0xlO n , 5.5xl0 n , 5.2xlO n , 5.1xl0 n , 5.05xl0 n , or 5.01xl0 n virus particles. In some embodiments, the therapeutic solution comprises 1.0-7.0xlO n or 1.0-5.5xl0 n virus particles. In some embodiments, the therapeutic solution comprises 4.5 -5.5x10 11 virus particles. In some embodiments, the therapeutic solution comprises 4.8-5.2xlO n virus particles. In some embodiments, the therapeutic solution comprises 4.9-5. lxlO 11 virus particles. In some embodiments, the therapeutic solution comprises 4.95-5.05xlO n virus particles. In some embodiments, the therapeutic solution comprises 4.99-5.01 x 10 11 virus particles In some embodiments, the kit further comprises an immunogenic component. In some embodiments, the immunogenic component comprises a cytokine selected from the group of IFN-γ, TNFa IL-2, IL-8, IL-12, IL-18, IL-7, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13. In some embodiments, the immunogenic component is selected from the group consisting of IL-7, a nucleic acid encoding IL-7, a protein with substantial identity to IL-7, and a nucleic acid encoding a protein with substantial identity to IL-7.

Kits

[00434] The compositions, immunotherapy or vaccines may be supplied in the form of a kit. The kits may further comprise instructions regarding the dosage and or administration including treatment regimen information.

[00435] In some embodiments, kits comprise the compositions and methods for providing combination multi -targeted cancer immunotherapy. In some embodiments, kits comprise the compositions and methods for the combination multi -targeted treatment of an infectious disease. In some embodiment's kits may further comprise components useful in administering the kit components and instructions on how to prepare the components. In some embodiments, the kit can further comprise software for conducting monitoring patient before and after treatment with appropriate laboratory tests, or communicating results and patient data with medical staff.

[00436] The components comprising the kit may be in dry or liquid form. If they are in dry form, the kit may include a solution to solubilize the dried material. The kit may also include transfer factor in liquid or dry form. If the transfer factor is in dry form, the kit will include a solution to solubilize the transfer factor. The kit may also include containers for mixing and preparing the components. The kit may also include instrument for assisting with the administration such for example needles, tubing, applicator, inhalant, syringe, pipette, forceps, measured spoon, eye dropper or any such medically approved delivery vehicle. In some embodiments, the kits or drug delivery systems as described herein also include a means for containing compositions disclosed herein in close confinement for commercial sale and distribution. EXAMPLES

EXAMPLE 1: Peptides and Vectors

[00437] The following HLA-A2 and HLA-A24 binding peptides were used in this and other examples: (a) the HLA-A2 binding CEA agonist peptide CAP1-6D (YLSGADLNL), (b) the HLA- A2 MUC1 agonist peptide P93L (ALWGQDVTSV), (c) the HLA-A24 binding MUC1 agonist peptide C6A (KYHPMSEYAL), and (d) the HLA-A2 binding brachyury agonist peptide

(WLLPGTSTV). All peptides were greater than 96% pure.

[00438] Ad5 [E1-, E2b-] -brachyury, Ad5 [E1-, E2b-]-CEA and Ad5 [E1-, E2b-]-MUCl were constructed and produced. Briefly, the transgenes were sub-cloned into the El region of the Ad5 [E1-, E2b-] vector using a homologous recombination-based approach. The replication deficient virus was propagated in the E.C7 packaging cell line, CsCl 2 purified, and titered. Viral infectious titer was determined as plaque-forming units (PFUs) on an E.C7 cell monolayer. The VP concentration was determined by sodium dodecyl sulfate (SDS) disruption and spectrophotometry at 260 nm and 280 nm. The CEA transgene also contained a modified CEA containing the highly immunogenic epitope CAP1-6D.

[00439] The sequence encoding for the human Brachyury protein (T, NM 003181.3) was modified by introducing the enhancer T-cell HLA-A2 epitope (WLLPGTSTV; SEQ ID NO:22) and removal of a 25 amino acid fragment involved in DNA binding. The resulting construct was subsequently subcloned into the Ad5 vector to generate the Ad5 [E1-, E2b-] -Brachyury construct.

[00440] The MUC1 molecule consisted of two regions: the N-terminus (MUCl-n), which is the large extracellular domain of MUC1, and the C-terminus (MUCl-c), which has three regions: a small extracellular domain, a single transmembrane domain, and a cytoplasmic tail. The cytoplasmic tail contained sites for interaction with signaling proteins and acts as an oncogene and a driver of cancer motility, invasiveness and metastasis. For construction of the Ad5 [E1-, E2b-]-MUCl, the entire MUC1 transgene, including eight agonist epitopes, was subcloned into the Ad5 vector. The agonist epitopes included in the Ad5 [E1-, E2b-]-MUCl vector bind to HLA-A2 (epitope P93L in the N-terminus, VIA and V2A in the VNTR region, and CIA, C2A and C3A in the C-terminus), HLA-A3 (epitope C5A), and HLA-A24 (epitope C6A in the C-terminus). The Tri-Ad5 vaccine was produced by combining of 10 10 VP of Ad5 [E1-, E2b-] -Brachyury, Ad5 [E1-, E2b-]-CEA and Ad5 [E1-, E2b-]-MUCl at a ratio of 1 : 1 : 1 (3x10 10 VP total).

EXAMPLE 2: Multiple Injections of Ad5Null Adenovirus Vector Produces Anti-Adenovirus Antibodies

[00441] This example shows that multiple injections of Ad5-null results in the production of anti- adenovirus antibodies in the injected subjects.

[00442] It was demonstrated that the Ad5-null adenovirus vector that does not contain any heterologous nucleic acid sequences, generated a neutralizing immune response in mice. In one experiment, female Balb/c mice aged 5-7 weeks were immunized with Ad5Null viral particles at 14 day intervals. To determine the presence of anti-adenovirus antibodies, an enzyme linked immunosorbent assay (ELISA) was used. For this ELISA, 10 9 viral particles were coated onto microtiter wells in 100 μΐ ^ of 0.05M carbonate/bicarbonate buffer, pH 9.6, and incubated overnight at room temperature. For a standard immunoglobulin G (IgG) reference curve, 200 ng, 100 ng, 50 ng, 25 ng, and 0 ng of purified mouse IgG were coated onto microtiter wells as described above. After incubation, all wells were washed 3 times with 250 μΐ ^ of 1% bovine serum albumin (BSA) in phosphate buffered saline (PBS), pH 7.4. After washing, 250 μΐ ^ of BSA/PBS was added to all and incubated for 30 minutes at room temperature to block unbound sites. After incubation, all wells were washed 3 times with 250 μΐ ^ of BSA/PBS. After washing, 200 μΐ ^ of a 1/100 serum dilution in BSA/PBS was added to wells and incubated for 1 hour at room temperature. For a positive control, 200 μΐ ^ of a 1/10000 dilution of anti-adenovirus antiserum in BSA/PBS was added to wells. Control wells contained BSA/PBS only. After incubation, all wells were washed 3 times with 250 μΐ ^ of BSA/PBS. After washing, 200 μΐ ^ of a 1/10000 dilution of peroxidase conjugated γ-chain specific goat anti-mouse IgG (Sigma Chemicals) in BSA/PBS were added to each well and incubated for 1 hour at room temperature. After incubation, all wells were washed 3 times with 250 μΐ ^ of

BSA/PBS. After washing, 200 μΐ ^ of developing reagent (0.5 mg/mL 1,2-phenylene-diamine in 0.2M potassium phosphate buffer, pH 5.0, containing 0.06% hydrogen peroxide) was added to each well and incubated for 30-40 minutes at room temperature. After incubation, the color reaction was stopped by addition of 50 μΐ ^ 5N HC1 to each well. All wells were then read in a microwell plate reader at 492 nm. After readings were obtained, the optical density readings of unknown samples were correlated with the standard IgG curve to obtain the ngs of IgG bound per well. This was performed using the INSTAT statistical package.

ELISA to detect antibodies against CEA

[00443] ELISA plates were coated with 100 ng of human CEA (Sigma- Aldnch) in 0.05 M carbonate-bicarbonate buffer pH 9.6 and incubated overnight at room temperature. Plates were washed three times with phosphate buffered saline containing 1% Tween-20 (PBS-T) and then blocked with PBS containing 1% BSA for 60 min at room temperature. After an additional three washes, serum diluted 1/50 in PBS-T was added to the wells and the plates were incubated for 1 hour at room temperature. Peroxidase labeled goat anti-mouse immunoglobulin (Ig) G (γ-chain specific) (Sigma- Aldrich) antibody at a 1 :5000 dilution was added to the wells after washings and plates were incubated for 1 hour. Plates were washed three times and 1,2-phenylene-diamine substrate solution was added to each well. The reaction was stopped by adding 10% phosphoric acid. Absorbance was measured at 492 nm on a SpectraMax 190 ELISA reader. The nanogram

equivalents of IgG bound to CEA per well was obtained by reference to a standard curve generated using purified mouse IgG and developed at the same time as the CEA ELISA. The results were analyzed and quantitated using SoftMax Pro 6.3 software.

[00444] Significant levels (P<0.001) of anti-adenovirus IgG antibody were detected in mice 2 weeks after a first injection with 10 10 Ad-5-null (FIG. 1). A significantly higher level (P<0.001) was observed 2 weeks after a second injection with 10 10 adenovirus. Significantly higher (P<0.001) levels of antibody were continued to be observed 2 weeks after a third injection with 10 10 Ad5-null. Each value represents the average of triplicate determinations from pooled sera of 5 mice in each group. Multiple injections of Ad5-null resulted in production of anti-adenovirus antibodies in the subjects.

[00445] To determine the presence of neutralizing antibody to Ad, the following assay was utilized. A HEK-293T-cell line was cultured in 200 μΕ of culture medium consisting of DMEM containing 10% fetal calf serum (DMEM/FCS) in micro well tissue culture plates at a cell concentration of 2x10 3 cells per well for 24 hours at 37 °C in 5% CO 2 . After incubation, 100 μΕ of culture medium was removed from triplicate wells and mixed with 20 μΕ of DMEM/FCS containing viral particles (VP). After mixing, the 120 μΕ mixture was added back to the respective micro wells. In another set of triplicate wells, 100 μΕ of culture medium was removed and mixed with 20 μΕ of heat inactivated (56 °C for 1 h) Ad immune mouse serum previously incubated with VP for one hour at room temperature. After mixing, the 120 μΕ mixture was added back to the respective wells. In triplicate cell control wells, 20 μΕ of DMEM/FCS was added to control for total culture medium volume. Triplicate medium-only control wells contained 220 μΕ of DMEM/FCS. The tissue culture plate was incubated for an additional 3 days at 37 °C in 5% CO 2 . After incubation, 40 μΕ of PROMEGA cell viability reagent (Owen's reagent) was added to all wells and incubated for 75 minutes at 37 C in 5% C0 2 . In this assay, the Owen's reagent (MTS tetrazolium compound) is bioreduced by viable cells into a colored formazan product that is soluble in tissue culture medium. The quantity of formazan product as measured by absorbance at 490 nm is directly proportional to the number of living cells in culture. After incubation, 150 μΕ was removed from each well and transferred to another microwell plate for optical density readings. Optical density readings at 492 nm were subsequently obtained using a microwell plate reader.

[00446] To detect the presence of neutralizing antibodies to Ad, groups of 5 mice each were injected once, twice, or three times with 10 10 Ad5-null at two week intervals. Two weeks after the final injection of virus, mice were bled, pooled, and assessed for neutralizing antibody as described above using 4x10 7 VP incubated with or without heat inactivated sera. Cells cultured alone served as a control group. Normal mice and mice injected one time with Ad5null did not exhibit significant levels of neutralizing antibody (FIG. 2). Mice injected two times with Ad exhibited significant (P<0.05) levels of neutralizing antibody as compared with cells incubated with virus only. Mice injected three times with Ad5-null also exhibited significant (P<0.01) levels of neutralizing antibody as compared with cells incubated with virus only.

EXAMPLE 3: The Ad5 [El-J-CEA Vector Vaccine Induces CEA Specific Immune Response Upon Re-Immunization in Ad5 Immune Mice

[00447] This example shows that the Ad5 [E1-, E2b-] vector platform induces CMI responses against the tumor associated antigen (TAA) carcinoembryonic antigen (CEA) in the presence of preexisting Ad5 immunity in mice.

Characterization of Ad5 CEA Vectors

[00448] Initial studies were performed to confirm CEA gene expression of two Ad5-CEA vector platforms. It was first determined that the CEA antigen could be expressed on cells transfected with the vaccine vector platforms. A549 cells were obtained from ATCC and transfected with Ad5 [El -]- CEA or Ad5 [E1-, E2b-]-CEA. Western blot analysis revealed that cells transfected with the vector platforms expressed CEA antigen. (FIG. 35)

Methods

[00449] A549 cells were inoculated at a MOI of 555 VPs/cell with Ad5 [El -, E2b-]-CEA. Cells were incubated for 48 hours at 37°C in 5% C0 2. After 48 hours cells were harvested and washed with PBS and freeze/thawed three times. The whole cell lysate was heated for 70 °C for 10 min prior to loading on the gel. Recombinant CEA control was loaded at 30ng/Lane and the prepared lysate at 20 μΕ/lane. Sample loading buffer was included as an additional negative control and the positive controls were Magic Mark CP Western markers and the recombinant CEA. The gel was transferred to a nitrocellulose membrane and blocked with SuperBlock Blocking solution for 60 min. The membrane was probed with mouse monoclonal anti-CEA primary antibody (1 : 1000) and a secondary anti-mouse HRP (1 :2500) conjugated antibody. The membrane was washed three times then incubated with SuperSignal chemiluminescent reagent and banding was visualized by exposing X-ray film to the membrane followed by development.

Induction of Ad5 Immunity in Mice

[00450] To assess the levels of Ad5 immunity that could be induced, groups of Ad5 naive C57B1/6 mice were injected subcutaneously with the Ad5 vector platform (VP). Twenty eight to forty two days later, serum samples were collected and assessed for endpoint Ad5 NAb titers. As shown in FIG. 3, undetectable Ad5 NAb titers (endpoint Ad5 NAb titer < 1/25) were observed in normal control mice. Ad5 NAb (endpoint titers of 1/25 to 1/50) was detectable after one injection but dramatically increased after three injections of 10 10 Ad5. Therefore, in additional Ad5 immune studies, mice were injected twice with 10 10 Ad5 VP to render the animals Ad5 immune.

Immunization of Ad5 Immune Mice with Ad5 [El-]-CEA or Ad5 [E1-, E2b-1-CEA.

[00451] These experiments were designed to determine and compare the immunization induction potential of Ad5 [El-]-CEA and Ad5 [E1-, E2b-]-CEA vaccines in Ad5 immune mice. Groups of female C57B1/6 mice, 4 to 8 weeks old, were immunized 2 times at 2 week intervals with 10 10 Ad5- null VP. Two weeks following the last Ad5-null immunization, the mice were immunized 3 times at weekly intervals with 10 10 VP of Ad5 [El-]-CEA or Ad5 [E1-, E2b-]-CEA. Two weeks following the last immunization, mice were euthanized and their spleens and sera harvested for analyses.

[00452] CMI responses were assessed by ELISpot assays performed on splenocytes exposed to intact CEA antigen. Splenocytes from Ad5 immune C57B1/6 mice that were immunized

subcutaneously with Ad5 El-]-CEA or Ad5 [E1-, E2b-]-CEA were harvested and assessed for the number of IFN-γ and IL-2 secreting cells as described above. Significantly elevated numbers of both IFN-γ and IL-2 secreting cells were observed in spleens assayed from mice immunized with Ad5 [E1-, E2b-]-CEA as compared to immunized Ad5 [El-]-CEA mice (FIG. 4A and FIG. 4B).

Specificity studies revealed that immunizations with Ad5 CEA vectors induced specific CEA associated CMI responses and not responses against other irrelevant antigens such as the FflV-gag protein or β-galactosidase. These results demonstrate that immunization of Ad5 immune mice with Ad5 [E1-, E2b-]-CEA induce significantly higher CMI responses.

Lack of Adverse Liver Effects in Immunized Mice

[00453] Toxicity studies were performed on serum from Ad5 immune female C57B1/6 mice immunized with Ad5 [El-]-CEA, Ad5 [E1-, E2b-]-CEA as described above. Ad5 naive or Ad5 immune mice injected with buffer alone served as controls. Three days after the third immunization, aspartate aminotransferase (AST) levels were assessed on the blood samples to determine liver toxicity due to the treatment. AST levels were not elevated over controls following immunization with either vector (FIG. 5). Alanine aminotransferase (ALT) levels were also assessed and similar results were observed.

Ad5 [E1-, E2b-]-CEA Immunotherapy in Ad5 Immune Tumor Bearing Mice

[00454] Based upon the successful immunological results observed above, studies in which MC38 tumors were established in mice and then treated were performed as described below. For these studies a CEA expressing MC38 murine cell line was used. This cell line has been genetically modified to express human CEA and can be implanted into C57B1/6 mice. After tumor

establishment, the mice were treated with the novel Ad5 [E1 -, E2b-]-CEA vector platform. To determine if Ad5 immune tumor bearing mice could be treated with the Ad5 [E1-, E2b-]-CEA vector, C57B1/6 mice were injected two times subcutaneously with 10 10 Ad5 [El-]-null VP at 14 day intervals to render the mice Ad5 immune. Two weeks after the last injection, two groups of 7 C57B1/6 mice were injected subcutaneously with 10 6 CEA expressing MC38 tumor cells. Seven days later, when tumors were palpable, one group of mice was treated by distal subcutaneous injection with 10 10 VP of Ad5 [E1-, E2b-]-CEA on days 7, 13 and 19. A group of 7 injection buffer only treated C57B1/6 mice served as untreated controls. All mice were monitored for tumor size over a 21 day period and tumor volumes were determined as previously described. [00455] The tumor growth by day 19 was significantly reduced in the Ad5 [E1-, E2b-]-CEA treated mice and remained so (FIG. 6). At the end of the study (Day 22), the mice were sacrificed and the tumors were excised and weighed. Tumor measurements were taken and volumes were determined. Statistical analysis was performed using the Bonferroni post -tests analysis with PRISM software. The tumors in the mice treated with Ad5 [E1-, E2b-]-CEA were significantly (P<0.05) smaller in weight than the untreated controls (FIG. 7).

[00456] At the termination of the study, spleens were collected from mice and the CEA specific CMI response was determined by ELISpot assay. CEA specific IFN-γ secretion response was significantly higher in mice immunized with Ad5 [E1-, E2b-]-CEA than in mice who received MC- 38 tumor cells alone. These results indicate that treatment of CEA expressing tumors in Ad5 immunized mice using the Ad5 [E1-, E2b-]-CEA vaccine can significantly decrease tumor growth progression.

EXAMPLE 4: Quantitative ELISA for CEA expression on A549 cells after infection

[00457] This example shows a dose response evaluation using the Ad5 [E1-, E2b-] CEA vector to transduce the human cancerous lung cell line, A-549. The results show that the CEA antigen can be expressed in a dose dependent manner.

Experimental Design

[00458] On day one, of the assay a BD Falcon Tissue Culture 96-well plate was seeded with A549 cells passaged three days prior (lot# 30Jul02, passage p+23), (7.7xl0 3 cells/well) and placed into a 37±2 °C incubator with a 5±2% C0 2 atmosphere overnight.

[00459] The next day, a dilution series of the test article were prepared and replicate wells were inoculated at levels ranging from 1.56xl0 3 to 2.5xl0 4 viral particles/well. Untreated A549 cells were used to serve as the mock sample. On day four of the assay wells were treated with a 10% Triton X- 100 solution for analysis by ELISA to measure CEA concentration. _For the ELISA, a microtiter plate was coated overnight with an anti-CEA capture antibody (abeam Carcino embryonic antigen CEA antibody [(NCRC16(AKA161))]). The wells were washed to remove unbound reactants, and the plate was blocked with a Phosphate Buffered Saline (PBS) solution containing 1% Tween 20 to fall within the range of the standard curve. After the blocking period, the wells were washed, and samples, controls, and standards were incubated in assigned triplicate wells. Unbound reactants were removed by washing, and a rabbit polyclonal antibody to a CEA detection antibody was added. After incubation, the wells were washed and incubated with 3,3',5,5'-tetramethylbenzidine (TMB), the peroxidase substrate. The substrate formed a colored product in the presence of the enzyme, reaction was stopped with 1 M phosphoric acid solution, and the absorbance was determined on a calibrated microplate reader. A calibration curve was generated from standards containing known

concentrations of CEA, and the curve was used to determine the concentration of CEA in the samples._The quantity of CEA produced per virus particle was calculated from the concentration of CEA measured by ELISA, after adjusting for dilution and multiplicity of infection (MOI). The value determined in a similar manner for culture media alone was subtracted to compensate for background levels present in the media. The sample analysis is shown in Table 2.

TABLE 2

EXAMPLE 5: Schedule, dose, route of immunization safety data

[00460] Initial pre-clinical studies were performed to evaluate and confirm that an Ad5 [El -, E2b-] vector platform could express the antigen proteins on transfected cells. A-549 cells were transfected with vaccine platforms and analyzed by Western Blot Analysis (FIG. 8). Antigen proteins such as HTV-gag, FflV-pol, or FflV-nef were observed to be expressed on cells once they were transfected with the Ad5 [E1-, E2b-] vector platforms.

[00461] A dose response evaluation was performed using the Ad5 [E1 -, E2b-] vector platform and demonstrated that 10 10 virus particles (VP) is a dose that results in a desired CMI response against a transgene product in a murine model. CMI responses were assessed by utilizing an ELISpot assay to detect interferon-γ (IFN-γ) and IL-2 secreting cells (splenocytes) from spleens of mice. Furthermore, in murine and non -human primate (NHP) models, three immunizations using 10 10 VP separated by two weeks to four weeks, respectively, resulted in the desired CMI responses. In mice, a greater degree of CMI responses were observed after multiple immunizations as compared with one immunization only (FIG. 9). [00462] In a NHP model, the animals were rendered Ad5 immune by injection with wild type Ad5 virus. When Ad5 neutralizing antibody titers reached 1 :50 or greater, which confirmed that the animals were immune to Ad5, they were immunized intradermally three times at 30 day intervals with Ad5-[E1-, E2b-]-gag at a dose of lxlO 10 VP. 32 days after last vaccination and 124 days after the first immunization (wild type Ad5), the NAb titers were equal to or greater than 1: 1000. After immunizations, the presence of robust CMI responses was detected, when peripheral blood mononuclear cells (PBMCs) of animals were assessed for IFN-γ and IL-2 secreting cells (FIG. 10).

[00463] In addition to the preliminary immunology studies performed in the initial vaccine trial in 3 NHP shown above, toxicity studies were also performed on the same NHP vaccinated with Ad5 [E1-, E2b-]-HTV gag. Animal temperatures and weights were assessed during the study period. The animals gained weight as they grew during the study period. No temperature differences were observed during the study period. Hematology studies were also performed on the vaccinated NHP. There appeared to be a small increase in the white blood cell count 2 weeks after the second vaccination that normalized thereafter.

[00464] Other than fluctuation in values, there appeared to be no other differences in hematology values during the course of the study. Chemistry values were also determined in the NHP during the course of the study. Alkaline phosphatase levels declined slightly during the course of the study but remained in the normal range. Albumin levels declined slightly during the course of the study but remained in the normal. There were no other differences observed in the blood chemistries during the course of the study. The route of immunization in this clinical study is chosen since the preponderance of DCs reside in the dermis.

[00465] A desired level of CMI response was induced using the Ad5 [E1-, E2b-] platform employing CEA and other transgenes. Using an Ad5 [E1-, E2b-]-CEA vector platform, both non- Ad5 immune and Ad5 pre-immunized mice were injected three times with the vaccine. After immunizations, the splenocytes from mice were assessed by ELISpot for IFN-γ secreting cells. Elevated CMI responses were observed after immunizations and the levels of CMI responses were similar in both non-Ad5 immune and Ad5 pre-immunized mice (FIG. 11). These results indicate that robust CMI responses can be induced despite the presence of pre-existing Ad5 immunity. A III clinical study was designed using three immunizations separated by three weeks via a needle subcutaneous delivery method.

Rationale for schedule, dose, route of administration

[00466] A clinical study design flowed from pre-clinical and clinical studies in animals and humans using the Ad5 [E1-, E2b-] vector platform. A dose response evaluation using the Ad5 [E1 -, E2b-] vector platform was performed demonstrating that 10 10 VPs is a dose which results in a desired CMI response against a transgene product in a murine model. Furthermore, in murine and non-human primate (NHP) models three immunizations using 10 10 VP separated by two to four weeks respectively resulted in the desired CMI. The route of immunization is chosen since a preponderance of dendritic cells (DCs) reside in the dermis. Using this premise, multiple murine and NHP studies were performed using a subcutaneous injection protocol. A desired level of circulating CMI was induced using the Ad5 [E1-, E2b-] platform employing CEA and other transgenes. A phase ΠΙ clinical study followed using three immunizations separated by three weeks via a needle

subcutaneous (SQ) delivery method continuing immunotherapy treatment every three months until removed from study for any reason including death.

Summary

[00467] The cDNA sequence containing the modified CEA with the CAP1(6D) mutation was produced. Clinical grade Ad5 [E1-, E2b-]-CEA(6D) was constructed and manufactured using the E.C7 cell line. A total of 34 patients (32 colorectal cancer patients, one bladder cancer patient, and one lung cancer patient) were entered into the Phase I/II clinical study under IND14325. The majority received all three scheduled immunotherapy treatments with Ad5 [E1-, E2b-]-CEA(6D). Five patients who stopped immunotherapy early did so due to significant disease progression.

RECIST 1.0 criteria using CT or MRI scans obtained at baseline and after treatments were completed. Toxicity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Peripheral blood CEA levels, hematology, serum chemistries, and anti-nuclear antibody titers were compared between baseline and 9 weeks following the initiation of immunotherapy. Survival was measured from the day of the first immunization until death from any cause.

[00468] A total of 94 treatments were administered to patients. No dose limiting toxicity or serious adverse events (SAE) that resulted in treatment discontinuation at any treatment dose level. There was only one significant change in a blood hematology value. As a group, the basophil count was significantly lower at week 9, three weeks after treatment ended. However, this value remained in the normal range for basophil counts and, overall, there appeared to be no significant biological effect. With a median follow-up of 7.4 months, all 34 patients as a group (cohorts 1, 2, 3/phase II, and cohort 5) experienced a 12-month survival proportion of 41.4%. Of the 34 patients entered into the study, 28 patients received the three immunotherapy treatments and experienced a 12-month survival proportion of 55%. For the colorectal adenocarcinoma patients, 27 patients received the three immunotherapy treatments and experienced a 12-month survival proportion of 53%. A dose response to increasing levels of Ad5 [E1-, E2b-]-CEA(6D) was observed with the highest cell- mediated immune (CMI) responses occurring in patients that received the highest dose of 5x10 11 VP of Ad5 [E1-, E2b-]-CEA(6D). When the highest CEA specific CMI responses were compared with pre-existing or vector induced Ad5 NAb activity, there was no correlation between levels of CEA specific CMI and Ad5 neutralizing antibody (NAb) level. These clinical trial data lead us to believe that there is sufficient data to advance to a randomized Phase ΠΙ trial for the treatment of metastatic colorectal adenocarcinoma with overall survival as the clinical endpoint.

Protocol schema and patient treatment.

[00469] The clinical study was performed under an FDA-approved Investigational New Drug Exemption and registered at ClinicalTrials.gov. Participants were recruited from medical oncology clinics at Duke University Medical Center (NC) and Medical Oncology Associates (WA). Patients provided informed consent approved by the respective Institutional Review Boards (IRB). Eligibility requirements included metastatic cancer expressing CEA and adequate hematologic, renal, and hepatic function. Trial participants were required to have received treatment with standard therapy known to have a possible overall survival benefit or refused such therapy. Exclusion criteria included chemotherapy or radiation within the prior 4 weeks, history of autoimmune disease, viral hepatitis, HTV, or use of immunosuppressive agents. Patients who had been receiving bevacizumab or cetuximab for at least 3 months prior to enrollment were permitted to continue receiving these antibodies. Prior CEA immunotherapy was permitted. The study employed a standard 3 + 3 dose escalation strategy with dose limiting toxicities (DLT) defined as grade 3 or 4 major organ toxicity. The Ad5 [E1-, E2b-]-CEA(6D) doses were delivered to patients as follows: cohort 1 : dose of lxl 0 9 VP in 0.5 rriL subcutaneously (SQ) in the same thigh every 3 weeks for 3 immunizations.; cohort 2: dose of 1X10 10 VP in 0.5 mL SQ every 3 weeks for 3 treatments; cohort 3: dose of lxlO 11 in 0.5 mL SQ every 3 weeks for 3 treatments. Following establishment of the dose of 1x10 11 VP as safe, an additional 12 patients received Ad5 [E1-, E2b-]-CEA(6D) at this dose and schedule (phase Π cohort). After completing the phase II cohort, an additional cohort (cohort 5) of six (6) patients received a dose of 5xl0 n VP in 2.5 mL SQ every 3 weeks for 3 treatments to determine safety of the highest achievable dose. PMBC were collected from patients just prior to the immunizations at weeks 0, 3, 6, and three weeks following the last treatment. The PBMC were frozen in liquid nitrogen until ELISpot assays were performed. In cohort 5, fresh PBMC were analyzed in preliminary flow cytometry assays for polyfunctional CD8+ T lymphocytes.

Assessment of clinical activity.

[00470] Clinical activity was assessed according to Response Evaluation Criteria in Solid Tumors (RECIST 1.0 criteria) using computed tomography (CT) or magnetic resonance imaging (MRI) scans obtained at baseline and after treatments were completed. Toxicity was assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 4. Peripheral blood CEA levels, hematology, serum chemistries, and anti-nuclear antibody titers were compared at baseline and 3 weeks following the final treatment. Survival was measured from the day of the first immunization until death from any cause

Analysis of CMI responses by ELISpot Assay. [00471] An ELISpot assay for IFN-γ secreting lymphocytes was performed. Briefly, isolated PBMCs (2x10 5 cells/well) from individual patient samples were incubated 36-40 h with a CEA peptide pool (15mers with 11 aa overlap covering full length CEA with the 6D modification; 0.1 μg/well) to stimulate IFN-γ producing T-cells. CMI responses to Ad5 were determined after exposure of patient PBMC to Ad5-null (empty vector). Cells stimulated with concanavalin A (Con A) at a concentration of 0.25 μg/well served as positive controls. Colored spot-forming cells (SFC) were counted using an Immunospot ELISpot plate reader and responses were considered to be positive if 50 SFC were detected/10 6 cells after subtraction of the negative control and SFC were >2- fold higher than those in the negative control wells

Determination of Ad5 neutralizing antibody (NAb) titers.

[00472] Endpoint Ad5 NAb titers were determined. Briefly, dilutions of heat inactivated test sera in 100 μΕ of DMEM containing 10% fetal calf serum were mixed with 4x10 7 VP of Ad5 [El -]-null and incubated for 60 minutes at room temperature. The samples were added to microwells containing HEK293 cells cultured in DMEM containing 10% heat inactivated calf serum at 2x10 3 cells/well for 24 hours at 37° C in 5% CO 2 . The mixture was incubated for an additional 72 hours at 37 °C in 5% C0 2 . An MTS tetrazolium bioreduction assay was used to measure cell killing and endpoint Ad5 NAb titers. Endpoint titers with a value less than 1 :25 were assigned a value of 0.

Statistics

[00473] Statistical analyses comparing immune responses were performed employing the Mann- Whitney test (PRISM, Graph Pad). Survival comparisons were performed employing Kaplan-Meier plots (PRISM, Graph Pad). Ad5 NAb titer and CEA-specific CMI were analyzed as continuous variables. The association of Ad5 NAb titer with change in CEA-specific CMI was tested with the Spearman correlation coefficient. The association of Ad5 NAb titer with survival was tested with the Wald test of the proportional hazards model. All tests used a 2-sided a of 0.05.

Demographics: All Patients.

[00474] Thirty two patients with metastatic colorectal cancer, one with lung cancer and one with bladder cancer, median age 58 (range 38-77), who had failed a median of three prior

chemotherapeutic regimens (range: 2->5), had a median performance status of 90% (range 70%- 100%), and had a median of three sites of metastatic disease (range 1 -5), were enrolled (Table 3). The majority of patients were able to receive all three immunizations. Five patients who stopped immunizations prior to completion of all three treatments did so due to significant disease progression. Patient demographics are shown in Table 3. TABLE 3

Dx = diagnos s (Bl= bladder cancer; C= colon cancer; L= lung cancer; R= rectal cancer)

KPS=Karnofsky Performance Status *concurrent cetuximab; Concurrent bevacizumab; @ concurrent panitumumab

++ Represents disease status at 9 weeks post-initiation of immunizations

PD = Progressive Disease; SD = Stable Disease

(+) Alive; (-) Dead at last follow-up; survival rounded off to nearest month

Demographics: Colorectal Adenocarcinoma Patients

[00475] Thirty two patients, median age 57.5 (range 38-77) with metastatic colorectal cancer, who had failed a median of three prior chemotherapeutic regimens (range: 2->5), had a median performance status of 90% (range 70%- 100%), and had a median of three sites of metastatic disease (range 1-5), were enrolled (Table 2). The majority was able to receive all three immunizations. Four patients who stopped immunizations early did so due to significant disease progression. The colorectal adenocarcinoma patient demographic compares favorably with previously published studies of patients with chemotherapy-refractory colorectal cancer.

Adverse Effects

[00476] A total of 94 immunization treatments were administered to all patients. There was no dose limiting toxicity and no serious adverse events that resulted in treatment discontinuation at any vaccine dose level. The most common toxicity (Table 3) was a self-limited, injection site reaction. Other reactions that occurred at a low frequency include fever, flu-like symptoms, anorexia, chills, nausea, and headache. These symptoms were also self-limiting and did not require intervention other than symptomatic measures such as acetaminophen.

Summary of Hematology. Chemistry, and ANA values pre and post treatment

[00477] Biological effects of Ad5 [E1-, E2b-]-CEA(6D) injections were monitored by recording blood hematology, chemistry, and anti -nuclear antibody (ANA) values of individual patients in case record forms (CRFs). Of 34 total patients entered into the trial, 28 received all three treatments with Ad5 [E1-, E2b-]-CEA(6D). For the 28 patients which received all three treatments, the blood hematology, chemistry, and ANA values at week 0 (prior to first treatment) were compared with those obtained at week 9 (three weeks after the third treatment). As shown in Table 4 below, there were no significant changes in chemistry or ANA values after treatments with Ad5 [E1 -, E2b-]- CEA(6D). There was only one significant change in the blood hematology values. The basophil count was significantly (P=0.0403) lower at week 9 after treatments. However, this value remained in the normal range for basophil counts and overall there were no significant biological effects. Clinical Outcomes

[00478] CEA levels at baseline and week 9 were assessed in patients. Among those with CEA levels available at baseline and follow-up, three (patients 010, 020, and 024) had no increase in CEA levels at the end of the immunization period while the remaining patients showed increased CEA levels. There were three patients with stable disease who remained so during the 9 week study period. All other patients experienced some level of progressive disease (Table 2). Of the seven patients in cohorts 1 and 2, there were five deaths and two patients remained alive at 12 months following the initiation of immunization. Of the 21 patients in cohort 3 and phase II, there were 10 deaths and all the remaining 11 patients were alive at 12 months, respectively. Of the six patients in cohort 5, there were three deaths and three patients were alive at 6, 7, and 8 months, respectively.

[00479] Of the 34 patients enrolled into the study, two patients received one treatment, four patients received two immunization treatments, and the remaining 28 patients received all three immunization treatments. All patients were followed for survival and Kaplan-Meier plots and survival proportions performed (PRISM software). Patient deaths were determined by information gathered from the social security death index (SSDI) database and clinical charts.

[00480] The seven patients in cohorts 1 and 2 experienced a 12-month survival proportion of 29% (FIG. 12A). Of the patients in cohorts 1 and 2, patient 004 survived 11 months and received additional post-immunization treatments with bevacizumab, folfox, and xeloda. Patient 003 survived nine months and received irradiation treatment after Ad5 [E1-, E2b-]-CEA(6D)immunizations. Patient 005, alive at 12+ months, received irradiation treatment and entered another clinical trial after immunizations. Patient 010 survived up to 12 months and entered two clinical trials after immunizations. Patients 002, 007, and 008 received no further treatments after immunizations and survived 3, 1, and 12+ months, respectively.

[00481] The 21 patients in cohort 3 and phase Π experienced a 12-month survival proportion of 48% (FIG. 12B). Of the patients in cohort 3 and phase II, one patient (017) received concurrent cetuximab during immunizations. Patients 020 and 021 received concurrent bevacizumab during immunizations. Patient 011 surviving over 12 months received radiation treatment after Ad5 [E1-, E2b-]-CEA(6D) immunizations. Patients 012 and 016 survived over 12 months and 6 months, respectively, and received additional chemotherapy treatment after immunizations. Patient 013 survived 4 months and received treatment with nexavar after immunizations. Patient 015 survived 10 months and received follow-on treatment with cetuximab. Patient 019 survived over 12 months and received treatment with bevacizumab and xeliri after protocol immunizations. Patient 020 survived over 12 months and received treatment with bevacizumab after immunizations. Patient 021 survived over 12 months and received follow-on treatment with bevacizumab and xeloda. Patient 500 survived over 12 months and received treatment with cetuximab and xeloda and entered a clinical trial after immunizations. Patient 501 survived over 12 months and received treatment with cetuximab and irinotecan after Ad5 [E1-, E2b-]-CEA(6D) immunizations. Patient 508 has survived over 12 months; however, further data on the characteristics of this patient was unable to be obtained. Patients 017, 023, 024, 025, 026, 502, 504, 506, and 507 received no further treatment after immunizations and survived 3, 4, 12+, 7, 4, 3, 12+, 3, and 12+ months, respectively (+ means still alive at the time of writing).

[00482] The six patients in cohort 5 experienced a 12-month survival proportion of 50% (FIG. 12C). Of the patients in this cohort, one patient (030) is currently alive at 8 months and received treatment with pazopanib and threshold 302 chemotherapy after Ad5 [E1-, E2b-]-CEA(6D) immunizations. Patient 031 is currently alive at 7 months and has not received further treatment after immunizations. Patient 032 received concurrent panitumumab, survived 6 months, and received treatment with folfox after immunizations. Patient 033 received concurrent panitumumab, survived 5 months with no additional therapy after immunizations. Patient 034 is currently alive at 6+ months and received radiation and treatment with xeloda after immunizations. Patient 035 received two treatments and survived 2 months.

[00483] With a median survival of 7.4 months, all 34 patients as a group (cohorts 1, 2, 3/phase II, and cohort 5) experienced a 12-month survival proportion of 41% (FIG. 12D). Of the 34 patients entered in to the study, 28 patients received the three immunization treatments and experienced a 12- month survival proportion of 55% (FIG. 13A) with a median survival of 10.625 months. For the colorectal adenocarcinoma patients, 27 patients received the three immunization treatments and experienced a 12-month survival proportion of 53% (FIG. 13B) with a median survival of 10.00 months.

Evaluation of immune parameters in treated metastatic colorectal cancer patients

[00484] A secondary objective was to evaluate CEA specific immune responses following immunization treatments with the product.

[00485] Dendritic cells were generated from the peripheral blood mononuclear cells (PBMCs) of a prostate cancer patient (HLA-A2 + and -A24 + ) enrolled in a clinical trial employing a PSA-TRICOM vaccine in combination with ipilimumab; using PBMCs from this patient post-vaccination, individual T-cell lines specific for CEA, MUC1, and Brachyury were unable to be established.

Briefly, PBMCs were isolated using lymphocyte separation medium gradient, resuspended in ΑΓΜ- V medium (2x10 7 cells) and allowed to adhere in a 6-well plate for 2 hours. Adherent cells were cultured for 5 days in AIM-V medium containing lOOng/ml of recombinant human (rh) GM-CSF and 20ng/ml of rhIL-4. The culture medium was replenished every 3 days.

[00486] As determined by ELISA, no antibody activity directed against CEA was observed. CMI responses in colorectal cancer patients treated in cohort 1, cohort 2, cohort 3/Phase II, and cohort 5 were assessed. PBMCs were isolated prior to Ad5 [E1-, E2b-]-CEA(6D) treatment and after all treatments as well as three weeks following the last treatment from patients. CEA specific ELISpot assays were performed on PBMCs to determine the numbers of interferon γ (IFN-γ) secreting lymphocytes after exposure to CEA peptides in vitro. The highest CMI responses during

immunizations were determined, regardless of time point (weeks 3, 6, or 9) in the patients treated in cohort 1, cohort 2, cohort 3/Phase II, and cohort 5. This analysis revealed a dose response to increasing levels of product. The highest CMI levels occurred in patients that received the highest dose of 5xl0 n VP (Cohort 5) (FIG. 14).

Determination of Induced CMI Responses to CEA.

[00487] ELISpot analysis was performed on cryopreserved PBMC samples drawn before each immunization and after completion of the final immunization to assess CEA-specific CMI responses. A dose response effect with the highest magnitude CEA-specific CMI responses occurring in patients who received the highest dose of Ad5 [E1-, E2b-]-CEA(6D) was observed(FIG. 14). Of the doses received, 0/3 (0%) patients in cohort 1 exhibited positive CEA-directed CMI responses, 1/4 (25%) patients in cohort 2 exhibited positive CEA-directed CMI responses, 10/19 (53%) patients in cohort 3/phase II exhibited positive CEA-directed CMI responses, and 4/6 (67%) patients in cohort 5 exhibited positive CEA-directed CMI responses. The time course of induction of CEA-specific CMI (FIG. 15) demonstrated that there may be plateau in the magnitude of CEA CMI prior to the last dose. In the largest group of patients who received the same dose (cohort 3 plus phase II), a significant increase over baseline in the average CEA-directed CMI responses at the 6 week evaluation (P<0.05, Mann-Whitney test) was observed, averaging 94 SFC/106 PBMC, which increased further by the 9 week evaluation (FIG. 15). One patient (patient ID 13) had a highly elevated baseline CEA-specific immune response (1100 SFC) and had elevated CMI at week six (2305 SFC) but did not return for 9-week evaluation and was not included in CEA CMI analysis.

[00488] Ad5 NAb and CMI against Ad5 was also measured and correlated with CEA-specific CMI. Each patient had their serum and PBMC sample tested at baseline (prior to treatment) and at 9 weeks after completion of 3 treatments. Nineteen of 31 colorectal cancer patients (61%) tested in this study had Ad5 neutralizing activity in serum samples prior to the onset of treatment with Ad5 [El -, E2b-]-CEA(6D). The mean pre-treatment Ad5 NAb titer value obtained among all patients was 1 : 189 ±1 :71 SEM (geometric mean 1 :21) and the mean pre-treatment Ad5 NAb titer among seropositive patients was 1 :308 ± 1 : 108 (geometric mean 1 : 146). Analysis of serum samples from patients who received 3 immunizations revealed Ad5 NAb titers that were significantly increased (P<0.0001, Mann-Whitney test) by week 9 (mean 1 :4767 ± 1 : 1225 SEM (geometric mean 1: 1541) when compared with their respective baseline values (FIG. 16). Analysis of PBMC for CMI responses to Ad5 also revealed a significant increase (P<0.01, Mann-Whitney test) in Ad5 directed CMI responses after immunizations with Ad5 [E1-, E2b-]-CEA(6D) (FIG. 16).

[00489] Comparison of week 9 CEA-directed CMI responses from patients with low baseline preexisting Ad5 immunity (Ad5 NAb >200) verses those with high baseline Ad5 immunity (Ad5 NAb>200) revealed no significant difference in immune responses (P>0.4, Mann Whitney test) (FIG. 17). Further, when the highest CEA specific CMI responses were compared with pre-existing or vector induced Ad5 NAb activity, there was no correlation between levels of CEA CMI and Ad5 NAb activity (FIG. 17). These data indicate that immunizations with Ad5 [E1-, E2b-]-CEA(6D) were not only able to overcome self-tolerance, but were also able to induce CEA-specific immune responses in colorectal cancer patients despite the presence of pre-existing and/or immunization induced Ad5 immunity. Together these clinical trial data support the advancement to a Phase ΠΙ clinical trial with overall survival as the primary endpoint.

Adverse Effects, Hematology, Chemistry, and ANA values

[00490] In this Phase l/ll trial, the Ad5 [El -, E2b-]-CEA(6D) was demonstrated to be suitable to be manufactured to scale, as well be easily and repeatedly administered by conventional subcutaneous injection techniques. The most common adverse effects were site of injection reactions and flu-like symptoms consisting of fever, chills, headache, and nausea. There was no impact on blood hematology or serum chemistries and, overall, the treatments were well tolerated. Specifically, no SAE were noted, and no treatments were stopped due to adverse events, indicating that a dose limitation to use of Ad5 [E1-, E2b-]-CEA(6D) in this clinical application had not been met.

[00491] These data suggest that patients with advanced colorectal cancer which are treated with Ad5 [E1-, E2b-]-CEA(6D) do not have serious adverse effects and may experience extension of life even if they have pre-existing immunity to Ad5. The results of this trial were encouraging enough to advance to a large, randomized, single agent trial. The observation that some of the patients experienced an increase of CMI which is dose dependent, indicates that this may play a role in their clinical outcome.

Table 4. Adverse Events

Parenthesis ( ) indicates numbers of events.

Based on 94treatments

Values represent inverse of the titer and are from patients with positive values.

Discussion

[00492] Adenoviral vectors have significant potential for use as cancer therapeutic vaccines because of their propensity to induce robust adaptive immune responses specifically against transgene products in general. However, recombinant first generation Ad5 [E1-] vectors used in homologous prime/boost regimens have been greatly limited in their potential efficacy due to the presence of pre-existing Ad5 immunity as well as vector induced immunity. Specifically, Ad5- directed immunity mitigates immune responses to TAA that have been incorporated into earlier generation Ad5 [E1-] based platforms. The Ad5 [E1-, E2b-] platform utilized in the present study was intended to accommodate a homologous prime-boost regimen, by avoiding presentation of antigens that are the targets of pre-existing Ad5 immunity.

[00493] CEA was investigated as a transgene to be incorporated into the new Ad5 [El -, E2b-] vector platform for use as a cancer therapeutic vaccine. CEA expression in adults is normally limited to low levels in the gastrointestinal epithelium, whereas, CEA is over-expressed in adenocarcinomas of the colon and rectum and in many breast, lung, and pancreas cancers. The HLA A2 restricted CAP1(6D) modification of CEA was chosen because compared with the wild type CAPl epitope, CAP1(6D) can enhance the sensitization of CTLs and has been included in recent CEA-based vaccine constructs. Although HLA type was not tested for because a full length CEA was used that is not HLA-restricted, A*0201 is the allele observed most frequently in Caucasians (allele frequency 0.2717) and is common in other populations. However, it is possible to test patients for HLA type and utilize the relationship between HLA type and clinical and/or CMI responses.

[00494] Multiple subcutaneous immunizations employing three administrations of a single dose level (lxlO 10 VP) of this class of Ad5 vaccine expressing the TAA CEA, (Ad5 [E1-, E2b-]- CEA(6D)) were tested in a pre-clinical murine model of CEA expressing cancer. In mice with preexisting Ad5 immunity, the induction of potent CEA directed CMI responses were demonstrated that resulted in anti-tumor activity and noted that these CMI and anti-tumor responses were significantly greater than those responses induced by a current generation Ad5 [E1-] based vector vaccine. In additional animal models (both cancer and infectious disease targeted) it was demonstrated that multiple subcutaneous immunizations with vaccines based on the new Ad5 [E1-, E2b-] platform induce CMI responses that were superior to those of current generation Ad5 [E1-] based vaccines, can overcome the barrier of Ad5 immunity, and can be utilized in multiple immunization regimens requiring a generation of robust CMI responses. The greatest magnitude of CEA-directed CMI responses occurred in patients receiving the highest dose of the vector. A CEA-directed CMI response was induced in a dose-responsive manner despite the presence of pre-existing and/or vector induced Ad5 immunity. No CEA directed antibody responses were observed either pre- or post- vaccination employing an ELISA technique. A population of polyfunctional CD8+ T-cells (those that secrete more than one cytokine when activated) after immunizations were also observed, a sign of greater functionality of T-cells induced by the vaccine. These data support the use of the Ad5 [El , E2b-]-CEA(6D) vector in homologous prime-boost regimens designed to induce and increase CEA-directed CMI responses in patients with advanced colorectal adenocarcinoma, as well as any number of other vaccine amenable diseases or applications.

[00495] As compared to earlier generation Ad5 [E1-] vectors containing deletion in the early 1 (El) gene region, the Ad5 [E1-, E2b-] vector platform with additional deletions in the early 2b (E2b) gene region exhibits significantly reduced inflammatory responses directed at the vector. This can result in longer transgene expression and a reduction in elimination of transgene expressing cells (e.g., antigen presenting cells) that would otherwise occur due to induced inflammatory responses. Since Ad5 late gene antigen expression is significantly reduced as compared to earlier generation Ad5 platforms, this could enable the Ad5 [E1-, E2b-] platform to evade Ad5 immune mediated neutralizing activity for significantly longer periods of time resulting in greater longevity and amplification of TAA expression. In addition, an E2b gene product, a polymerase, is a target of human cellular memory immune responses to Ad5 infection and its elimination from the vaccine could be furthering its capability in the setting of pre-existing Ad5 immunity. Without being bound by theory, the extended and/or greater expression of TAA by the vector in this milieu could result in a more effective immune response against the target antigen. However, it is also possible that this vector configuration produces better transgene expression, different biodistribution, or different innate/adaptive immune effects that impact the effectiveness of this vector, rather than escape from pre-existing immunity.

[00496] Of interest is the observation that treated patients in this study exhibited favorable survival probability. Overall, all 25 patients treated at least 2 times with Ad5 [E1 -, E2b-]-CEA(6D) exhibited a 12 -month survival probability of 48% and this was achieved despite the presence of significant levels of pre-existing Ad5 neutralizing antibody titers. Without being bound by theory, by engaging the patient's immune system, active immunotherapeutics, such as Ad5 [E1-, E2b-]-CEA(6D), could induce continuous immunologic anti-tumor responses over a long period of time that could result in a "deceleration" or alteration in specific aspects of the rapid growth rate or spread of the tumor not measured by standard response assessments. Indeed, slower tumor progression in Ad5 immune mice harboring established CEA-expressing tumors following treatment with Ad5 [E1-, E2b-]-CEA(6D) were observed. Moreover, it has been noted that overall survival might be the only true parameter for determination of clinical efficacy of any potential cancer (immune) therapy.

EXAMPLE 6: Clinical study of CEA (6D) immunotherapeutic in metastatic colorectal cancer

[00497] Phase MI clinical trial in mCRC using Ad5 [E1-, E2b-]-CEA(6D): The objectives of this first in man phase I/II dosing trial were to assess safety, evaluate CEA-specific immune responses in mCRC patients, and obtain data on overall survival. The trial was performed under FDA-approved IND14325 and registered at ClinicalTnals.gov (NCTO 1147965). The Ad5 [E1-, E2b-]-CEA(6D) doses were administered subcutaneous (sc) every 3 weeks for 3 treatments as follows: Cohort 1 (3 patients) received 10 9 VP each treatment; Cohort 2 (3 patients) received 10 10 VP; Cohorts 3/4 (21 patients) received 10 11 VP; and Cohort 5 (5 patients) received 5xlO n VP.

Patient Demographics:

[00498] Thirty-two mCRC patients with CEA expressing cancers were enrolled into the study. The 32 mCRC patients enrolled had a median age 57.5 (range 38-77), had failed a median of three prior chemotherapeutic regimens (range: 2-5), had a median performance status of 90% (range 70%- 100%), and had a median of three sites of metastatic disease (range 1 -5). The majority of patients received all three immunizations. Patients who stopped immunizations early did so due to significant disease progression. The mCRC demographics compared favorably with previously published studies with chemotherapy-refractory mCRC. Adverse Effects:

[00499] There were no dose-limiting toxicities and no serious adverse events that resulted in treatment discontinuation at any dose level. The most common toxicity was a self-limited, injection site reaction. The blood hematology, chemistry, and anti -nuclear antibody (ANA) values at week 0 (before the first treatment) were compared with those obtained at week 9. There were no biologically relevant changes in chemistry, hematology, or ANA values.

Clinical Outcomes:

[00500] Patients were washed out from anti -cancer treatments for 30 days prior to immunotherapy. Both Kras wild type and mutated patients were enrolled.

[00501] The mCRC patients were followed for long-term survival and Kaplan-Meier survival plots performed (PRISM software). Events were determined from the social security death index database and clinical charts. Median overall survival was 11 months and overall survival was 30% at 18 months and 20% at 29 months (FIG. 18).

Immune Responses:

[00502] A secondary objective was to evaluate CEA-specific immune responses during immunotherapy. Again, since the mCRC patients represented the highest number of patients treated, immunogenicity studies were performed on them. As determined by ELISA there was no detectable antibody activity directed against CEA in serum samples.

[00503] CMI responses of mCRC patients treated in all cohorts were assessed. Peripheral blood mononuclear cells (PBMC) were isolated from patients before and after each immunotherapy treatment, as well as three weeks after the last treatment. CEA-specific ELISpot assays were performed on PBMC as previously described to determine the number of IFN-y-secreting cells (SFC) after exposure to CEA peptides. The highest CMI responses during immunizations, regardless of time point (weeks 3, 6, or 9) were determined. This analysis revealed a dose-response relationship with increasing levels of vaccine (FIG. 19); the highest CMI levels occurred in patients who received the highest dose of 5xl0 n VP (Cohort 5) and the responses were observed despite the presence of pre-existing Ad5 immunity in a majority (61%) of patients. Analysis of serial PBMC samples over time indicated that CEA directed CMI responses were induced and increased over the course of 3 immunizations (FIG. 20).

[00504] Briefly, CMI (IFN-γ secretion) was assessed at baseline (Pre) and after administrations of Ad5 [E1-, E2b-]-CEA(6D) (Post). The highest CMI responses (regardless of time point) observed in the patients after treatment revealed a dose response. The highest CMI levels occurred in patients receiving the highest dose (Cohort 5) and was significantly elevated (P <0.02; Mann -Whitney test). Specificity of the responses was demonstrated by lack of reactivity with irrelevant antigens β- galactosidase and FflV-gag. For positive controls, PBMCs were exposed to concanavalin A. Values = Mean ± SEM (FIG. 19). [00505] Briefly, CMI (ELISpot IFN-γ SFC) responses in immunized mCRC patients were assessed at weeks 0, 3, 6, and 9. Note the increase in CMI responses during immunizations. Values=Mean ± SEM (FIG. 20).

Additional immune analyses on treated mCRC patients using flow cytometry.

[00506] Flow cytometry testing on PBMC from a patient with high CMI activity (>1000 ELISpot IFN-γ SFC) after exposure to CEA peptides revealed CD8+/IFN-Y+ cells (3.5%), CD8+/IFN- Y+/TNF-a+ cells (1.0%), CD4+/IFN-Y+ cells (0.4%), and CD4+/IFN-Y+/TNF-a+ cells (0.2%) demonstrating that polyfunctional cells were present.

[00507] Patient PBMC samples were also tested for HLA-A2 positivity by flow cytometry and 63% of the samples tested were HLA-A2 positive. When the highest CMI response level achieved per patient was assessed in association with the presence of HLA-A2, there was no significant difference observed between HLA-A2+ and HLA-A2- patients (264.6±119.0 IFN-γ SFC versus 165.6±108.1 IFN-γ SFC).

[00508] To assess the induction of cytolytic T lymphocyte (CTL) responses, ELISpot assays for granzyme B activity were performed. Granzyme B is a key mediator of target cell death via the granule-mediated pathway and the release of granzyme B by cytolytic lymphocytes upon effector- target interaction is an indicator of CTLs. The granzyme B ELISpot assay is a superior alternative to the 51Cr-release assay since it is significantly more sensitive and provides an estimation of cytotoxic effector cell frequency.

[00509] Increased granzyme B activity in PBMCs were observed after immunization (FIG. 16). Importantly, in an extended follow-up on PBMC samples from 5 mCRC patients, CMI responses were observed to decrease after immunizations (FIG. 22) were stopped indicating that further "booster" immunizations may be required to maintain induced CEA directed CMI activity.

[00510] Briefly, CTL responses (ELISpot granzyme B secreting SFC) were assessed pre (week 0) and post (week 6-9) treatment. Responses increased after immunizations (P < 0.05 i). Values = Mean ± SEM (FIG. 21).

[00511] Briefly, PBMC samples from 5 patients as assessed by ELISpot IFN-γ SFC. CMI responses peaked at week 9 and decreased by week 26 after treatment was stopped (FIG. 22).

[00512] These data indicate that multiple homologous immunizations with Ad5 [E1-, E2b-]- CEA(6D) induce CEA-specific CMI immune responses in patients despite pre-existing Ad5- neutralizing activity. These data further indicate that importantly, there was minimal toxicity and a favorable overall survival profile was observed. Finally, the results indicate that the novel Ad5 [El -, E2b-]-CEA(6D) gene delivery/expression platform can overcome tolerance to TAA and generate significant CMI responses to CEA in the setting of naturally acquired Ad5 -specific immunity. EXAMPLE 7: GLP Production of Clinical Grade Multi-targeted Vaccine

[00513] This example shows the production of clinical -grade multi-target vaccine using good laboratory practice (GLP) standards. Previously, the Ad5 [E1-, E2b-]-CEA(6D) product was produced using a 5 L Cell Bioreactor under GLP conditions in accordance with good manufacturing practice standards. This example shows that the Ad5 [E1-, E2b-]-mMUCl-C and the Ad5 [E1-, E2b- ]-Brachyury products can be produced in a 5 L Cell Bioreactor using a similar approaches.

[00514] Briefly, vials of the E.C7 manufacturing cell line will be thawed, transferred into a T225 flasks, and initially cultured at 37 °C in 5% C0 2 in DMEM containing 10% FBS/4 mM L-glutamine. After expansion, the E.C7 cells will be expanded using 10-layered CellSTACKS (CS-10) and transitioned to FreeStyle serum-free medium (SFM). The E.C7 cells will be cultured in SFM for 24 hours at 37 °C in 5% C0 2 to a target density of 5x10 5 cells/mL in the Cell Bioreactor. The E.C7 cells will then be infected with Ad5 [E1-, E2b-]-mMUCl-C or Ad5 [E1-, E2b-]-Brachyury, respectively, and cultured for 48 hours.

[00515] Mid-stream processing will be performed in an identical manner as that used to prepare clinical grade Ad5 [E1-, E2b-]-CEA(6D) product under IND14325. 30 minutes before harvest, Benzonase nuclease will be added to the culture to promote better cell pelleting for concentration. After pelleting by centrifugation, the supernatant will be discarded and the pellets re-suspended in Lysis Buffer containing 1% Polysorbate-20 for 90 minutes at room temperature. The lysate will then be treated with Benzonase and the reaction quenched by addition of 5M NaCl. The slurry will be centrifuged and the pellet discarded. The lysate will be clarified by filtration and subjected to a two- column ion exchange procedure.

[00516] To purify the vaccine products, a two-column anion exchange procedure will be performed. A first column will be packed with Q Sepharose XL resin, sanitized, and equilibrated with loading buffer. The clarified lysate will be loaded onto the column and washed with loading buffer. The vaccine product will be eluted and the main elution peak (eluate) containing Ad5 [E1 -, E2b-]-mMUCl-C or Ad5 [E1-, E2b-]-Brachyury carried forward to the next step. A second column will be packed with Source 15Q resin, sanitized, and equilibrated with loading buffer. The eluate from the first anion exchange column will be loaded onto the second column and the vaccine product eluted with a gradient starting at 100% Buffer A (20 mM Tris, 1 mM MgCl 2 , pH 8.0) running to 50% Buffer B (20 mM Tris, 1 mM MgCl 2 , 2M NaCl, pH 8.0). The elution peak containing Ad5 [El- , E2b-]-mMUCl-C or Ad5 [E1-, E2b-]-Brachyury will be collected and stored overnight at 2-8 °C. The peak elution fraction will be processed through a tangential flow filtration (TFF) system for concentration and diafiltration against formulation buffer (20 mM Tris, 25 mM NaCl, 2.5% (v/v) glycerol, pH 8.0). After processing, the final vaccine product will be sterile filtered, dispensed into aliquots, and stored at < -60 °C. A highly purified product approaching 100% purity is typically produced and similar results for these products are predicted. [00517] The concentration and total number of VP product produced will be determined spectrophotometrically. Product purity is assessed by HPLC. Infectious activity is determined by performing an Ad5 hexon-staining assay for infectious particles using kits.

[00518] Western blots will be performed using lysates from vector transfected A549 cells to verify mMUCl-C or Brachyury expression. Quality control tests will be performed to determine that the final vaccine products are mycoplasma-free, have no microbial bioburden, and exhibit endotoxin levels less than 2.5 endotoxin units (EU) per mL. To confirm immunogenicity, the individual vectors will tested in mice as described below (Example 8).

EXAMPLE 8: Immunogenicity of multi-targeted CEA, MUC1, T viral vector

[00519] This example shows immunogenicity results using the multi-targeted vaccine directed to

CEA, MUCl and T (Brachyury). Each viral vector product was tested for purity, infectivity, and antigen expression, as described herein and each passed these criteria.

Vaccination and splenocvte preparation

[00520] Female C57BL/6 mice (n=5) were injected s.c. with 10 10 VP of Ad5 [E1-, E2b-]- Brachyury or Ad5 [E1-, E2b-]-CEA or Ad5 [E1-, E2b-]-MUCl or a combination of 10 10 VP of all three viruses at a ratio of 1 : 1 : 1 (Tri-Ad5). Control mice were injected with 3 x 10 10 VP of Ad-null (no transgene insert). Doses were administered in 25 μΐ of injection buffer (20mM HEPES with 3% sucrose) and mice were vaccinated three times at 14-day intervals. Fourteen days after the final injection spleens and sera were collected. Sera were frozen at -20 C. Splenocyte suspensions were generated by gently crushing the spleens through a 70 uM nylon cell strainer (BD Falcon, San Jose, CA). Red cells were removed by the addition of red cell lysis buffer (Sigma- Aldrich, St. Louis, MO) and the splenocytes were washed twice and resuspended in R10 (RPMI 1640 supplemented with L- glutamine (2 mM), HEPES (20 mM), penicillin 100 U/ml and streptomycin 100 μg/ml, and 10% fetal bovine serum. Splenocytes were assayed for cytokine production by ELISPOT and flow cytometry.

Immunogenicity studies:

[00521] Previous studies assess induced immunity generated by the multi -targeted vaccine mixture. Immunization with Ad5 [E1-, E2b-] vectors is dose-dependent and lxlO 10 VP per dose was routinely used. Groups (N=5) of C57B1/6 mice were used.

[00522] This is study C57B1/6 mice were injected subcutaneously 3 times at 2-week intervals with tri-immunization comprising 3x10 10 viral particles (VP) Ad5 [E1-, E2b-]-null (empty vector controls) or with 3xl0 10 VP containing a 1 : 1: 1 mixture of Ad5 [E1-, E2b-]-CEA(6D), Ad5 [E1-, E2b-]-mMUCl-C, and Ad5 [E1-, E2b-] -Brachyury.

[00523] Two weeks after the last immunization CMI activity was determined employing ELISpot assays for IFN-γ secreting cells (SFC) after exposure of splenocytes to CEA, MUCl, or Brachyury peptide pools, respectively. [00524] Significant CMI responses to were detected in immunized mice (FIG. 23). Flow cytometry utilizing intracellular cytokine staining was performed on spleen cells after exposure to CEA peptides to assess the quantity of activated CD4+ and CD8+ T-cells.

[00525] Briefly, CMI responses against CEA, MUCl, and Brachyury as assessed by ELISpot assays for IFN-γ secreting splenocytes (SFC) were detected in multi -targeted immunized mice but not control mice (injected with Ad5-Null empty vector). Specificity of the ELISpot assay responses was confirmed by lack of reactivity to irrelevant SIV-nef or SIV-vif peptide antigens. A positive control included cells exposed to concanavalin A (Con A, data not shown). Values = Mean ± SEM (FIG. 23)

ELISpot assay

[00526] Brachyury-, CEA- and MUCl -specific IFN-γ- or IL-2-secreting T cells were determined by ELISpot assay from freshly isolated mouse splenocytes, as described above. Briefly, 2 xlO 5 splenocytes were stimulated with 0.2 μg/well of overlapping 15-mer peptides in a single pool derived from Brachyury or CEA, or MUCl. Cells were stimulated with Con A at a concentration of 0.0625 μg/per well as a positive control and overlapping 15-mer complete peptides pools derived from SIV-Nef and SrV-Vif were used as irrelevant peptide controls. The numbers of SFCs were determined using an Immunospot ELISpot plate reader and results were reported as the number of SFCs per 10 6 splenocytes.

[00527] TNF-a and IFN-γ expressing polyfunctional CD4+ and CD8+ cells were detected in immunized but not control splenocytes (FIG. 24). Testing was also performed on sera to detect induced CEA antibody using a previously described quantitative ELISA assay with purified CEA protein. Polyfunctional CD8+ (top) and CD4+ (bottom) cells expressing IFN-γ and TNF-a in mice immunized with the multi -targeted vaccine (right) but not in controls injected with Ad5-null (left). Specificity of the responses was confirmed by lack of reactivity to media alone or irrelevant SrV-nef or SIV-vif peptides. Values = Mean ± SEM (FIG. 24).

[00528] Significant antibody responses to CEA were detected in immunized but not control mice (FIG. 8). To determine the level of complement dependent cellular cytotoxicity (CDCC), a CDCC test was performed using murine MC38-CEA target cells. Significant CDCC activity was detected in sera of immune, but not control mice (injected with Ad5-null) (FIG. 25). Significant (P<0.0001) CEA antibody (left) and CDCC (right) responses were induced in mice immunized with the multi- targeted vaccine but not in control mice. Values = Mean ± SEM (FIG. 25).

Intracellular cytokine stimulation

[00529] Splenocytes were prepared as indicated for above. Stimulation assays were performed using lxl 0 6 live splenocytes per well in 96-well U-bottom plates. Pools of overlapping peptides spanning the entire coding sequences of Brachyury, CEA and MUCl were synthesized as 15-mers with 11 -amino acid overlaps and lyophilized peptide pools were dissolved in DMSO. Similarly constructed peptide pools corresponding to SIV-Vif and SIV-Nef served as off-target controls.

Splenocytes in RIO media (RPMI 1640, 10% fetal bovine serum, and antibiotics) were stimulated by the addition of peptide pools at 2 μg/mL/peptide for 6h at 37 °C and 5% C0 2 , with protein transport inhibitor (GolgiStop, BD) added 2h into the incubation. Stimulated splenocytes were then stained for lymphocyte surface markers CD8a and CD4, fixed, permeabilized, and then stained for the intracellular accumulation of IFN-γ and TNFa. Antibodies against mouse CD8a, CD4, IFN-γ, and TNFa were used and staining was performed in the presence of anti-CD16/CD32. Flow cytometry was performed and analyzed in BD Accuri C6 Software.

Complement-dependent cytotoxicity assay (CDC)

[00530] MC38-CEA2 tumor cells were cultured overnight at a density of 2x10 4 cells per well in 96- well tissue culture microplates. Pooled heat inactivated mouse sera were added at a 1 : 50 dilution and incubated at 37°C for 1 hour. Rabbit serum was then added at a 1 :50 dilution as a source of complement and cells were incubated an additional 2.5 hours at 37 °C. Cell culture supernatants were assayed using Promega Cytotox 96 non-radioactive cytotoxicity assay, according to the manufacturer's instructions. Percent lysis of MC38-CEA2 cells was calculated by the formula % lysis = (experimental - target spontaneous) / (target maximum - target spontaneous) x 100%.

Anti -tumor immunotherapy studies:

[00531] Studies were conducted to test the anti-tumor capability of Ad5 [E1-, E2b-]-based tri- vaccines in immunotherapy studies in mice with established CEA, MUCl, or Brachyury expressing tumors, respectively. In this study the anti-tumor activity of the individual components of the Ad5 [E1-, E2b-]-based tri-vaccine was assessed.

[00532] Groups (n=7) of C57B1/6 mice were injected subcutaneously in the right flank with 5xl0 5 CEA, MUCl, and/or Brachyury expressing murine tumor cells. After palpable tumors were detected, mice were treated by 3 subcutaneous injections with lxlO 10 VP each of Ad5 [E1-, E2b-]-null (no transgene, e.g., empty vector), Ad5 [E1 -, E2b-]-CEA(6D), Ad5 [E1-, E2b-]-mMUCl-C, and/or Ad5 [E1-, E2b-] -Brachyury, respectively. Tumor volumes were calculated and tumor growth curves were plotted. 7-10 mice/group are sufficient for statistical evaluation of treatment.

[00533] For in vivo tumor treatment studies, female C57BL/6 mice, 8-10 weeks old, were implanted with 10 6 MC38-MUC1 cells s.c. in the left flank. Mice were treated three times at a 7-day interval with 10 10 VP Adeno-MUCl or Tri-Ad5. Control mice were injected with 3 x 10 10 VP of Adeno-null. Tumor growth was assessed by measuring two opposing dimensions (a, b) and the volume calculated according to the formula V=(a x b) 2 /2 where the shorter dimension was "a". Tumor studies were terminated when tumors reached 1500 m 3 or became severely ulcerated.

[00534] Significant anti -tumor activity and increased survival in MUCl expressing tumor-bearing mice treated by immunotherapy (FIG. 26). Importantly, flow cytometry was used to determine that the MC38-MUC1 cell line expressed PDL1 and anti-tumor activity was achieved despite its presence. C57B1/6 mice (n=7/group) were inoculated with MC38-MUC1 expressing tumor cells subcutaneously in the left flank and administered 10 10 VP of Ad5-Null (empty vector) or 10 10 VP of Ad5 [E1-, E2b-]-mMUCl-C in the right flank on days 0, 7, 14.

[00535] Mice treated with Ad5 [E1-, E2b-]-mMUCl-C had significantly (P <0.05) smaller tumors on days 15 and 18 as compared to controls (top) and significantly longer survival (bottom). Values = Mean ± SEM. Experiment was terminated on day 36 (FIG. 26).

[00536] Immunotherapy of mice with Ad5 [E1-, E2b-]-Brachyury resulted in smaller tumors (FIG. 27). Briefly, C57B1/6 mice were inoculated with MC38-Brachyury expressing tumor cells subcutaneously in the left flank and administered 10 10 VP of Ad5-Null (empty vector) (N=4) or 10 10 VP of Ad5 [E1-, E2b-]-Brachyury (N=5) in the right flank on days 5, 11, and 17. Tumors were smaller in treated mice on days 15, 19, and 22. Values = Mean ± SEM (FIG. 27)

[00537] Larger numbers of mice will be treated to show significant anti-tumor activity and to combine immunotherapy with immune pathway checkpoint modulators, such as anti-checkpoint inhibitor antibodies, to determine if anti-tumor activity is enhanced.

EXAMPLE 9: In vitro validation of human T-cell activation by recombinant viral vectors

Tumor cell culture

[00538] Human colon carcinoma SW620 (HLA-A2 + , HLA-A24 + , Brachyury + , MUC1 + , CEA + ) and pancreatic carcinoma ASPC-1 (HLA-A1 + , HLA-A26 + , MUC1 + ) cell lines were used. Cell cultures were free of mycoplasma and maintained in complete medium (RPMI-1640 supplemented with 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin and 2 mM L-glutamine).

Detection of cytokines

[00539] Supernatants of T cells stimulated for 24 h with DCs infected with adenovirus vectors or pepti de-pulsed DCs in IL-2-free medium were evaluated for secretion of IFN-γ using an ELISA kit. The antigen-specific T-cell lines used in this analysis have been reported previously: (a) an HLA-A2 CEA-specific CTL, (b) an HLA-A2 MUC1 -specific CTL, (c) an HLA-A24 MUC1 -specific CTL, and (d) an HLA-A2 Brachyury-specific CTL.

Cytotoxic assay

[00540] In brief, target cells were labeled with 50 μθ of m In oxide at 37 C for 20 min and used at 3,000 cells/well in 96-well round-bottom culture plates. T cells were added at different ratios and incubated at 37 C for 16 h. Supernatants were harvested for gamma counting. Determinations were carried out in triplicate and SDs were calculated. Spontaneous release was determined by incubating target cells with medium alone and complete lysis was determined by incubating with 0.25% Triton X-100. Specific lysis was calculated with the use of the following formula: Lysis (%) = [observed release (CPM) - spontaneous release (CPM)] / [Complete release (CPM)- spontaneous release (CPM)] x 100. [00541] An in vitro study was performed to assess dendritic cells (DC) function and antigen- specific T-cell activation using the Ad5 [E1 -, E2b-]-CEA(6D), and Ad5 [E1-, E2b-]-Brachyury vectors. Briefly, human DC were cultured 6-7 days in media with IL-2 and GM-CSF. DC cultured in AFM-V medium were infected with Ad5 [E1 -, E2b-]-CEA(6D), or Ad5 [E1-, E2b-]-Brachyury, or Ad5 [E1-, E2b-]-null (empty vector) and incubated for 2 days. Uninfected DCs were used as controls.

[00542] Human DC cells were then analyzed by flow cytometry for expression of CD80 (a marker for DC maturation), CD83 (a marker for DC maturation), CD86 (a DC marker) and DR (a DC marker) as measured by mean fluorescence intensity. As shown in Table 6, infection of DC with Ad5 [E1-, E2b-]-based vectors induced activation and maturation of human DCs.

Table 6

[00543] Dendritic cells (2x10 ) in 1 mL of AIM-V medium were infected with adenovirus vectors (Ad5 [E1-, E2b-]-CEA, Ad5 [E1-, E2b-]-MUCl, Ad5 [E1-, E2b-]-Brachyury, and Ad5 [E1-, E2b-]- null at indicated multiplicity of infection (MOI of 10,000 or 20,000) for 1 hour in 6-well plates. AFM-V medium (4 mL) was then added to each well and incubated for an additional 2 days. To analyze the efficacy of transgene expression, DCs were harvested and analyzed using flow cytometry and Western blot. For phenotypic analysis, DCs were stained for the expression of CD80, CD83, CD86, CEA, and HLA-DR using BV421 -conjugated anti-CD80, PerCP Cy 5.5 -conjugated anti-CD83, APC-Cy7-conjugated anti-HLA-DR, PE-conjugated anti-CD86, and FITC-conjugated anti-CEA. As shown in Table 7, Ad5 vectors induced maturation and activation of human DCs. Indicated adenovirus vectors were used at a concentration of 10,000 or 20,000 multiplicity of infection (MOI) for infection of human dendritic cells (DCs). Expression of CD80, CD83, CD86 and FILA-DR were analyzed by flow cytometry. Results are expressed in % of positive cells (mean fluorescence intensity).

Table 7

Ad5 [E1-, E2b-]-CEA 10,000 56.8 (189) 48.7 (262) 99.2 (3877) 98.3 (6553)

Ad5 [E1-, E2b-]-CEA 20,000 54.5 (185) 46.6 (271) 99.1 (3628) 98.0 (6015)

Ad5 [E1-, E2b-]-MUCl 10,000 41.4 (167) 42.4 (251) 98.5 (3178) 96.9 (5227)

Ad5 [E1-, E2b-]-MUCl 20,000 46.7 (172) 44.3 (260) 98.9 (3591) 97.2(5779)

[00544] To determine if infected human DCs could stimulate human antigen-specific T-cell lines to secrete IFN-γ, infected DCs were incubated with antigen-specific T-cell lines and tested for IFN-γ secreting activity. As shown in Table 8, only the CEA or Brachyury antigen-specific HLA-A2+ T- cell lines incubated with respective HLA-A2+ DC infected with Ad5 [E1 -, E2b-]-CEA(6D) or Ad5 [E1-, E2b-] -Brachyury were stimulated to secrete IFN-γ. No IFN- γ secretion was detected in the controls. Infection of HLA-A2+ DC with Ad5 [E1-, E2b-]-based vectors can activate antigen- specific T-cells to produce IFN-γ.

Table 8

-, - - rac yury in ect e , ot one .

[00545] HLA-A2+ DC were infected with a mixture of Ad5 [E1 -, E2b-]-CEA(6D) and Ad5 [E1-, E2b-]-Brachyury. Infected DCs were used to generate specific cytotoxic HLA-A2+ T-lymphocytes (CTL) using autologous PBMC. The autologous DCs were used as APC for three in vitro

stimulations (IVSs). Autologous B cells pulsed with CEA or Bracyhyury were used to re-stimulate antigen-specific CTLs after the 3 rVSs. The effector T-cell to target tumor cell ratio was 30: 1 and the percent cytolytic activity was determined. As shown in Table 9, infection of HLA-A2+ DC with a mixture of Ad5 [E1-, E2b-]-CEA(6D) and Ad5 [E1-, E2b-] -Brachyury can generate antigen-specific cytolytic T-cells. Cytolytic activity was detected in an HLA-A2 dependent manner.

Table 9

[00546] To determine if infected human DCs could stimulate human antigen-specific T-cell lines to lyse target tumor cells. Human DCs (6-day culture in IL-4 and granulocyte-macrophage colony- stimulating factor (GM-CSF) 2x10 4 cells/well in 0.5 mL of AIM- V) were infected with indicated adenovirus vectors at 20,000 multiplicity of infection (MOI). After 48 hours, DCs were washed and used for stimulation of human antigen-specific T cells. Results are expressed in pg/ml of IFN-γ per lxlO 5 T cells/ml. Table 10 demonstrates that infection of human DCs with recombinant adenovirus vectors encoding CEA, MUC1, or Brachyury, can activate antigen specific T-cell lines. Numbers in bold indicate a significant enhancement of IFN-γ secretion compared to corresponding wells with uninfected DCs. [— indicates that the assay was not performed.]

[00547] Human DCs (6-day culture in IL-4 and GM-CSF) from an HLA-A2 and -A24 donor were infected with Tri-Ad5 vector at 2 x 10 4 /well (24-well plate) in 0.5 mL of AFM-V. Tri-Ad5 vectors were used at 20,000 MOI for 1 hour and then 1.5 mL of AIM-V were added to each well. Infected DCs were incubated for 48 hours and then washed and used for stimulation of human antigen- specific T cells. Results are expressed in pg of IFN-γ per 1 x 10 5 T cells/ml. Numbers in bold indicate a significant enhancement of IFN-γ secretion compared to corresponding wells with uninfected DCs.

[00548] CEA-, MUC1- and Brachyury-specific cytotoxic T lymphocytes (CTLs) were generated. Dendritic cells (1 -2x10 5 /well in 1 mL of AIM-V) were infected with 20,000 MOI of Tri-Ad5, as described above. Infected DCs were used as APCs for stimulation of autologous nonadherent cells at an effector-APC ratio of 10: 1. Cultures were incubated for 3 days at 37 °C in a humidified atmosphere containing 5% C0 2 . The cultures were then supplemented with rhIL-2 for 7 days; IL-2 containing medium was replenished every 3 days. The 10-day stimulation constituted one in vitro stimulation (IVS) cycle. Autologous vector-infected DCs were used as APCs for three IVS.

Autologous peptide-pulsed B cells were used to re-stimulate antigen-specific CTLs after three IVS. T-cell lines were maintained in medium containing IL-7 and IL-15 (10 ng/ml).

[00549] Human DCs from a prostate cancer patient (6-day culture in IL-4 and GM-CSF; 2x10 4 cells/well in 0.5 mL of AIM-V) were infected with Tri-Ad5 at 20,000 MOI. After 48 h, infected DCs were washed and used to generate specific cytotoxic T lymphocytes (CTLs) using autologous peripheral blood mononuclear cells (PBMCs) as effectors. Following 3 cycles of in vitro

stimulations, autologous peptides-pulsed B cells were used as antigen-presenting cells. Results are expressed in pg/ml of IFN-γ. [-- indicates that the assay was not performed.] As shown in Table 11, infection of human dendritic cells can generate antigen-specific T cells to Brachyury, MUC1 and CEA and produce IFN-γ when stimulated with autologous B cells pulsed with the peptides.

Table 11

[00550] CEA-, MUC1- and Brachyury-specific cytotoxic T lymphocytes (CTLs) were generated. Dendritic cells (1 -2x10 5 /well in 1 mL of AIM- V) were infected with 20,000 MOI of Tri-Ad5, as described above. Infected DCs were used as APCs for stimulation of autologous nonadherent cells at an effector- APC ratio of 10: 1. Cultures were incubated for 3 days at 37 °C in a humidified atmosphere containing 5% C0 2 . The cultures were then supplemented with rhIL-2 for 7 days; IL-2 containing medium was replenished every 3 days. The 10-day stimulation constituted one in vitro stimulation (TVS) cycle. Autologous vector-infected DCs were used as APCs for three IVS.

Autologous peptide-pulsed B cells were used to re-stimulate antigen-specific CTLs after three IVS. T-cell lines were maintained in medium containing IL-7 and IL-15 (10 ng/ml).

[00551] Human DCs (6-day culture in IL-4 and GM-CSF) from an HLA-A2 and -A24 donor were infected with Tri-Ad5 vector at 2 x 10 4 /well (24-well plate) in 0.5 mL of AIM-V. Tri-Ad5 vectors were used at 20,000 MOI for 1 hour and then 1.5 mL of AIM-V were added to each well. Infected DCs were incubated for 48 hours and then washed and used for stimulation of human antigen- specific T cells. Results are expressed in pg of IFN-γ per 1 x 10 5 T cells/ml. Infection of human dendritic cells with Tri-Ad5 vectors encoding transgenes can activate antigen-specific T cell lines to produce IFN-γ. As shown in Table 12, infection of human dendritic cells with Tri-Ad5 can generate antigen-specific T cells to Brachyury, MUC1 and CEA and produce IFN-γ when stimulated with autologous B cells pulsed with the corresponding peptides. Numbers in bold indicate a significant enhancement of IFN-γ secretion compared to corresponding wells with uninfected DCs. Table 12

[00552] These studies show that Ad5 [E1-, E2b-] -based vectors can induce maturation of human DCs in vitro, infected DCs can stimulate antigen-specific human T-cells, and infected DCs can generate antigen-specific human CTLs. In the studies reported here, multi-TAA targeted

immunotherapy (Tri-Ad5), which consisted of a mixture of three Ad5 vectors expressing different TAAs, is as efficient in the activation of human T cells as the use of each of the adenovirus vectors alone, with only minor differences. Nine different in vivo parameters were measured via vaccinating mice with each of the Ad5 [E1 -, E2b-]-CEA, Ad5 [E1-, E2b-]-MUCl, and Ad5 [E1-, E2b-]- Brachyury vectors individually versus vaccination with Tri-Ad5. Of the 21 assays performed, statistical differences observed were (a) an enhanced number of MUC1 -specific splenocytes and CD8 + IFN-producing and multifunctional CD8 + T cells, and (b) more CEA-specific CD8 + IFN- producing T cells, in the mice vaccinated with one vector than in the Tri-Ad5 vaccinated mice. On the other hand, the Tri-Ad5 vaccinated mice produced more CEA-specific IL-2-producing cells than the Ad5 [E1 -, E2b-]-CEA vaccinated mice. In the other 16/21 assays, however, there were no statistical differences in the results between the use of the individual vector versus the use of the Tri- Ad5 in terms of antigen-specific activation of (a) splenocytes for IFN-γ and IL-2 production, (b) CD8 + T cells for IFN-γ production, (c) CD4 + T cells for IFN-γ production, (d) multifunctional CD8 + T cells for IFN-γ and TNF-a production, (e) multifunctional CD4 + T cells for IFN-γ and TNF-a production, and (f) production of antigen-specific antibodies. There was also no difference in antitumor activity using a single vector (Ad5 [E1 -, E2b-]-MUCl) versus the Tri-Ad5 vaccine; while both vaccines did not eliminate the tumor, both vaccines reduced the tumor growth rate in a similar manner. While Tri-Ad5 was not as efficient in T-cell activation in some assays, the potential ability of the Tri-Ad5 platform to overcome the TAA heterogeneity that exists in human solid tumors far outweighs the relatively minor differences in potency of T-cell activation of Tri-Ad5 vs. individual vectors in some assays.

[00553] CEA, MUC1 and Brachyury are all human TAAs and are not expressed in murine solid tumors. Moreover, human solid tumors are very heterogeneous with respect to expression of different TAAs. It would be extremely difficult to transfect a murine tumor cell line with all three transgenes to define the effect of vaccination of Tri-Ad5 vs. each vector alone. The targeting of a murine tumor expressing MUC1 was chosen because the single Ad5 [E1-, E2b-]-MUCl vector was more potent in some of the murine T-cell assays compared to the Tri-Ad5 than the CEA and Brachyury vectors compared to Tri-Ad5. Thus this appeared to be the most stringent model to compare the Tri-Ad5 platform to a single vector platform. The studies reported herein were designed to provide the rationale for potential clinical studies as a vaccine immunotherapy, or use in combination with other therapeutics, using this novel adenovirus vaccine delivery platform (Ad5 [E1-, E2b-] targeting a diverse range of TAA transgenes in the Tri-Ad5 regimen.

[00554] While the checkpoint inhibitor antibodies have shown evidence of clinical activity in melanoma and squamous non-small cell lung cancer, clinical benefit in other cancer types has been observed in a minority of patients. For some tumor types, such as colorectal cancer and prostate cancer, the anti-PDLl/PDl checkpoint inhibitors have shown little clinical activity. One hypothesis that has been put forth for the lack of PDL1/PD1 therapeutic activity in some patients is the lack of T-cell infiltrates in tumors. Consequently, if a vaccine targeting TAAs in the tumor would result in the presence of antigen-specific T cells in the tumor microenvironment, then an immune pathway checkpoint modulator employed in combination or following vaccination would be able to "release the brakes" of the tumor-infiltrating anergized T cells leading to clinical effect.

EXAMPLE 10: Immunotherapy for HPV and ETBX-041 (Ad5 [E1-, E2b-]-HPV-E6/E7) Vaccine

[00555] This example shows that the combination therapy as provided herein reduces tumors and a murine model of HPV-associated cancers which express HPV early 6 (E6) and early 7 (E7) oncogenes. Briefly, tumor bearing HPV murine model mice were treated with the Ad5 [E1 -, E2b-]- HPV-E6/E7 vaccine comprising a modified non-oncogenic and fused HPV-E6/E7 gene and treated tumor bearing mice. The TC-1 murine tumor cell line used to induce tumors in the mice expressed HPV-E6/E7 as well as PDL1.

[00556] Flow cytometry was used to examine the tumors in HPV tumor bearing mice and calculated the abundance of various tumor infiltrating lymphocytes (TILs). The TILs in the tumors were observed to have the following cell types and abundance levels: myeloid derived suppressor cells (MDSC) (7.5% ± 2.4 SD), FoxP3 expressing T regulatory (Treg) lymphocytes (7.1% ± 2.5), PD1 expressing CD8a+ lymphocytes (25.5% ±16.4), and LAG-3 expressing CD8a+ lymphocytes (4.1% ± 2.3). This profile of TILs indicates that a suppressive immune pathway in present in the tumors.

[00557] The addition of immune checkpoint inhibitor antibodies, such as anti-PDl antibody, to the Ad5 [E1-, E2b-]-HPV-E6/E7 immunotherapy resulted in greater anti-tumor responses (FIG. 28). Additionally two mice in this group were observed to have essentially complete tumor regression (FIG. 29). Briefly, C57B1/6 mice (n=7/group) were implanted with HPV-E6/E7 expressing TC-1 tumor cells (day 0) and treated by immunotherapy on days 10, 17, 24 with 10 10 VP of Ad5-null (empty vector) plus 100 μg control IgG (intraperitoneal), 10 10 VP of Ad5-Null (empty vector) plus 100 μg anti-PDl, 10 10 VP of Ad5 [E1-, E2b-]-HPV-E6/E7 plus 100 μg mouse IgG, or 10 10 VP of Ad5 [E1-, E2b-]-HPV-E6/E7 plus 100 μg anti-PDl. Immunotherapy with or without anti-PDl resulted in significant inhibition of tumor growth by day 23 (P<0.05). All control mice were terminated by day 23 due to tumor mass. Values = Mean ± SEM (FIG. 28).

[00558] Tumor growth and regression in 2 mice treated by immunotherapy with anti-PDl injections were analyzed (FIG. 29). Tumor growth peaked on days 20 & 27, respectively, and then regressed thereafter. This study demonstrates that the combination of immune checkpoint inhibitor drugs in combination with use of multi -targeted vaccines can enhance anti-tumor effects.

[00559] Immunotherapy against human papilloma virus (HPV) using a viral gene delivery platform to immunize against HPV 16 genes E6 and E7 (Ad5 [E1-, E2b-]-E6/E7) combined with programmed death-ligand 1 (PD1) blockade was investigated to determine whether it could increase therapeutic effect as compared to the vaccine alone. Ad5 [E1-, E2b-]-E6/E7 as a single agent induced HPV- E6/E7 cell-mediated immunity. Immunotherapy using Ad5 [E1-, E2b-]-E6/E7 resulted in clearance of small tumors and an overall survival benefit in mice with larger established tumors. When immunotherapy was combined with immune checkpoint blockade, an increased level of anti -tumor activity against large tumors was observed. Analysis of the tumor microenvironment in Ad5 [E1 -, E2b-]-E6/E7 treated mice revealed elevated CD8 + tumor infiltrating lymphocytes (TILs); however, induction of suppressive mechanisms such as programmed death-ligand 1 (PDL1) expression on tumor cells and an increase in PD1 + TILs was seen. When Ad5 [E1-, E2b-]-E6/E7 immunotherapy was combined with anti-PDl antibody, CD8 + TILs were observed at the same level but a reduction in tumor PDL1 expression on tumor cells and reduced PD1 + TILs providing a mechanism by which combination therapy favors a tumor clearance state and a rationale for pairing antigen-specific vaccines with immune pathway checkpoint modulators in future clinical trials.

[00560] Herein, Ad5 [E1-, E2b-]-E6/E7 immunizations combined with PD1 blockade were examined for an increase an anti -tumor effect. Also, the CMI response induced by the Ad5 [E1-, E2b-]-E6/E7 vaccine was characterized and the kinetics of an anti -tumor response was determined to evaluate the therapeutic potential of treating small versus large established tumors. To investigate a possible mechanism of action, the relationship between the levels of effector T cells and suppressor T cells within the parenchyma of the tumor was evaluated and lymphocyte populations and expression of co-inhibitory molecules that may play a role in the observed anti-tumor responses were characterized.

Viral Construction

[00561] Ad5 [E1-, E2b-]-E6/E7 was constructed and produced. Briefly, the transgenes were sub- cloned into the Ad5 [E1-, E2b-] vector using a homologous recombination -based approach and the replication deficient virus was propagated in the E.C7 packaging cell line, CsCl 2 purified, and titered Viral infectious titer was determined as plaque forming units (PFU) on an E.C7 cell monolayer. The viral particle (VP) concentration was determined by sodium dodecyl sulfate (SDS) disruption and spectrophotometry at 260 nm and 280 nm. As a vector control, Ad5 [E1-, E2b-]-null was employed, which is the Ad5 platform backbone with no transgene insert.

Immunization and splenocyte preparation

[00562] Female C57BL/6 mice (n=5/group) were injected subcutaneously (SQ) with varying doses of Ad5 [E1-, E2b-]-E6/E7 or Ad5 [E1-, E2b-]-null. Doses were administered in 25μΕ injection buffer (20mM HEPES with 3% sucrose) and mice were immunized three times at 14-day intervals. Fourteen days after the final injection, spleens and sera were collected. Serum from mice was frozen at -20°C until evaluation. Suspensions of splenocytes were generated by disrupting the spleen capsule and gently pressing the contents through a 70 μηι nylon cell strainer. Red blood cells were lysed by the addition of red cell lysis buffer and after lysis, the splenocytes were washed twice in R10 (RPMI 1640 supplemented with L-glutamine (2mM), HEPES (20mM) (Corning, Corning, NY), penicillin (100 U/ml) and streptomycin (100 μg/mL), and 10% fetal bovine serum. Splenocytes were assayed for cytokine production by ELISpot and flow cytometry.

Enzyme-Linked Immunosorbent Spot (ELISpot) assay

[00563] HPV E6 and E7 specific interferon-γ (IFN-γ) secreting T cells were determined by

ELISpot assays using freshly isolated mouse splenocytes prepared as described above. The ELISpot assay was performed. Pools of overlapping peptides spanning the entire coding sequences of HPV E6 and E7 were synthesized as 15-mers with 11 -amino acid overlaps (and lyophilized peptide pools were dissolved in DMSO. Splenocytes (2x10 5 cells) were stimulated with 2 μg/mL/peptide of overlapping 15-mer peptides in pools derived from E6 or E7. Cells were stimulated with

Concanavalin A (Con A) at a concentration of 0.06 μg/per well as a positive control. Overlapping 15-mer complete peptide pools derived from SrV-Nef (AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH) were used as irrelevant peptide controls. The numbers of Spot Forming Cells (SFC) were determined using an Immunospot ELISpot plate reader (and results reported as the number of SFC per 10 6 splenocytes.

Intracellular Cytokine Stimulation

[00564] Splenocytes were prepared as described for the ELISpot assay above. Stimulation assays were performed using 10 6 live splenocytes per well in 96-well U-bottom plates. Splenocytes in R10 media were stimulated by the addition of HPV E6, HPV E7, or SrV-Nef peptide pools at

2μg/mL/peptide for 6 h at 37 °C in 5% CO 2 , with protein transport inhibitor (GolgiStop, BD) added two hours after initiation of incubation. Stimulated splenocytes were then stained for lymphocyte surface markers CD8a and CD4, fixed with paraformaldehyde, permeabilized, and stained for intracellular accumulation of IFN-γ and TNF-a. Fluorescent-conjugated antibodies against mouse CD8a (clone 53-6.7), CD4 (clone RM4-5), IFN-γ (clone XMG1.2), and TNF-a (clone MP6-XT22) were purchased from BD and staining was performed in the presence of anti-CD16/CD32 (clone 2.4G2). Flow cytometry was performed using an Accuri C6 Flow Cytometer (BD) and analyzed using BD Accuri C6 Software.

Tumor immunotherapy

[00565] For in vivo tumor immunotherapy studies, female C57BL/6 mice, 8-10 weeks old, were implanted with 2x10 5 TC-1 HPV-E6/E7 expressing tumor cells SQ in the left flank. Mice were treated three times at 7-day intervals with SQ injections of 10 10 VP Ad5 [E1-, E2b-]-E6/E7. Control mice were injected with 10 10 VP Ad5 [E1-, E2b-]-null under the same protocol. In combinational studies, mice were given 100 μg of rat anti-PDl (clone RMPl-14) or an isotype rat control antibody (clone 2A3) IP at the same time as immunization. Rat anti-PDl antibody and rat IgG 2a isotype control antibodies were purchased from BioXcell. Tumor size was measured by two opposing dimensions (a, b) and volume was calculated according to the formula V=(a 2 xb)/2 where a was the shorter dimension. Animals were euthanized when tumors reached 1500mm 3 or when tumors became ulcerated.

Analysis of tumor infiltrating cells (TILs) by flow cytometry

[00566] Four groups of 8-10 week old female C57BL/6 mice (n=5/group) were implanted with 2x10 5 TC-1 tumor cells SQ in the left flank at day 0. Two of these groups were immunized SQ with 10 10 VP Ad5 [E1-, E2b-]-null vector control and the other two groups SQ with 10 10 VP Ad5 [E1-, E2b-]-E6/E7 vaccine. These immunizations were administered twice at 7-day intervals starting on day 12. In addition to immunizations, mice in one Ad5 [E1-, E2b-]-E6/E7 group and one Ad5 [E1-, E2b-]-null group were administered 100μg rat anti-PDl (clone RMPl-14) SQ at days 12 and 16 and 100μg hamster anti-PDl (clone J43) at days 19 and 23 to increase the effective dose of anti-PDl. To control for treatment with these immune pathway checkpoint modulators, mice in the remaining Ad5 [E1-, E2b-]-E6/E7 and Ad5 [E1-, E2b-]-null groups were administered the relevant rat and hamster control IgG antibodies on the same days. Hamster anti-PDl antibody and isotype control were purchased from BioXcell. At day 27, tumors were measured, excised, and weighed. Tumors were minced and digested with a mixture of collagenase IV (lmg/ml), hyaluronidase (100μg/ml), and DNase IV (200U/ml) in Hank's Balanced Salt Solution (HBSS) at room temperature for 30 min and rotating at 80 rpm. Enzymes were purchased from Sigma- Aldrich. After digestion, the tumor suspension was placed through a 70 μιτι nylon cell strainer and centrifuged. Red cells were removed by the addition of red cell lysis buffer (Sigma- Aldrich) and after lysis, the tumor suspensions were washed twice in phosphate buffered saline (PBS) containing 1% (w/v) bovine serum albumin and resuspended in fluorescent activated cell sorting (FACS) buffer (PBS pH 7.2, 1% fetal bovine serum, and 2 mM EDTA) for staining. Fluorescent-conjugated antibodies against CD45 (30-F11), CD4 (RM4-5), and PDL1 (MIH5) were purchased from BD. Fluorescent-conjugated antibodies against CD8p (H35-17.2), CD25 (PC61.5), FoxP3 (FJK-16s), PD1 (RMP1-30), LAG- 3 (C9B7W), and CTLA4 (UC10-4B9) were all purchased from eBioscience. Surface staining was performed for 30 minutes at 4 °C in ΙΟΟμΙ. FACS buffer containing anti-CD16/CD32 (clone 2.4G2). Stained cells were washed in FACS buffer, fixed with paraformaldehyde, and (if needed) permeabilized in permeabilization buffer (eBioscience) before staining with fluorescent-conjugated anti-FoxP3 or anti-CTLA4 for 60 minutes at 4°C in ΙΟΟμΙ ^ permeabilization buffer containing anti-CD16/CD32 (clone 2.4G2). Cells were washed with permeabilization buffer, washed back into FACS buffer, and a fixed volume of each sample was analyzed by flow cytometry using a BD Accuri C6 flow cytometer. Tumor cells were defined as CD45 " events in a scatter gate that includes small and large cells. CD4 + TILs were defined as CD45 + /CD4 + events in a lymphocyte scatter gate. CD8 + TILs were defined as CD45 + /CD8p + events in a lymphocyte scatter gate. Regulatory T cells (Tregs) were defined as CD45 + /CD4 + /CD25 + /FoxP3 + events in a lymphocyte scatter gate. Effector CD4 + T cells were defined as CD45 + /CD4 + /CD25 FoxP3 " events in a lymphocyte scatter gate. Isotype-matched control antibodies were used to determine positive expression of FoxP3, PDL1, PD1, LAG-3, and CTLA4. Flow cytometry was performed using an Accuri C6 Flow Cytometer (BD) and analyzed in BD Accuri C6 Software.

HPV-E6/E7 specific cell mediated immune responses induced by Ad5 [E1-, E2b-]-E6/E7

[00567] A study was performed to determine the effect of increasing doses of Ad5 [E1-, E2b-]- E6/E7 immunizations on the induction of CMI responses in mice. Groups of C57BL/6 mice (n=5/group) were immunized SQ three times at 14-day intervals with 10 s , 10 9 , or 10 10 VP Ad5 [E1-, E2b-]-E6/E7. Control mice received 10 8 VP, 10 9 VP, or 10 10 VP Ad5 [E1-, E2b-]-null (empty vector controls). Two weeks after the last immunization, splenocyte CMI responses were assessed by ELISpot analysis for IFN-γ secreting cells. A dose effect was observed and the highest CMI response level was obtained by immunizations with 10 10 VP Ad5 [E1-, E2b-]-E6/E7 (FIG. 36). No responses were detected in control mice injected with Ad5

[E1-, E2b-]-null.

[00568] Intracellular accumulation of IFN-γ and TNF-a in both CD8a + and CD4 + splenocytes populations was also determined in mice immunized with 10 10 VP Ad5 [E1-, E2b-]-E6/E7.

Intracellular cytokine staining (ICS) after stimulation with overlapping peptide pools revealed E6 and E7 antigen-specific IFN-γ accumulation in CD8a + lymphocytes isolated from all mice immunized with Ad5 [E1-, E2b-]-E6/E7 (FIG. 37A). Peptide-stimulated splenocytes were also stained for the intracellular accumulation of TNF-a, and a significant population of multifunctional (IFN-y + /TNF-a + ) CD8a + splenocytes specific for both E6 and E7 were able to be detected (FIG. 37B)

Treatment of HPV-E6/E7 expressing tumors

[00569] The anti -tumor effect of immunotherapy treatment in mice bearing HPV-E6/E7 TC-1 tumors was investigated. These tumor cells expressed PDL1 as assessed by flow cytometry analysis. When labeled with PE-conjugated anti-PDLl, the TC-1 cells had a median fluorescent intensity (MFI) of 537 whereas cells labeled with a PE-conjugated isotype control antibody had an MFI of 184, demonstrating the presence of the immune suppressive PDL1 on the surface of the TC-1 cells. Two groups of C57BL/6 mice (n=5/group) were inoculated with 2xl0 5 TC-1 tumor cells SQ into the right subcostal area on day 0. On days 1, 8, and 14 mice were treated by SQ injections of 10 10 VP Ad5 [E1-, E2b-]-null (vector control) or 10 10 VP Ad5 [E1-, E2b-]-E6/E7. All mice were monitored for tumor size and tumor volumes were calculated. Mice immunized with Ad5 [El -, E2b-]-E6/E7 had significantly smaller tumors than control mice beginning on day 12 (p<0.01) and remained significantly smaller for the remainder of the experiment (p<0.02), including 3 of 5 mice showing complete tumor regression (FIG. 38A). Tumors in mice from the vector control treated group began reaching the threshold for euthanasia starting on day 26 and all mice in this group were euthanized by day 33, whereas mice in the Ad5 [E1-, E2b-]-E6/E7 treated group were all alive with complete tumor regression of small tumors (<150mm 3 ) at the end of experiment on day 36 (FIG. 38B).

[00570] To determine if immunotherapy with Ad5 [E1-, E2b-]-E6/E7 was effective against larger tumors, TC-1 tumor cells tumors were implanted in two groups of C57BL/6 mice (n=4/group) and then delayed weekly treatment with Ad5 [E1-, E2b-]-E6/E7 for 6 days post tumor implantation, at a time when tumors were small but palpable. Mice beginning treatment on day 6 initially

demonstrated tumor growth similar to the control group; however, beginning on day 16, tumor regression was observed (FIG. 39A). The tumors in mice that began treatment on day 6 were significantly smaller (p<0.05) than the control group beginning on day 20 and 3 of 4 mice had complete regression by day 27. Ad5 [E1-, E2b-]-E6/E7 administration beginning on day 6 also conferred a significant survival benefit (p<0.01) (FIG. 39B).

[00571] Finally, to determine if immunotherapy with Ad5 [E1-, E2b-]-E6/E7 was effective against large established tumors, TC-1 tumor cells were implanted in two groups of C57BL/6 mice

(n=4/group) then delayed weekly treatment with Ad5 [E1-, E2b-]-E6/E7 until 13 days post tumor implantation, when tumors were ~ 100mm 3 . In this treatment group, initial tumor growth was observed to be similar to the control group but some mice in the control group reached euthanasia criteria on day 23, preventing analysis of significance at further time points (FIG. 40A). However, tumor volumes in the Ad5 [El -, E2b-]-E6/E7 treated group were below the euthanasia threshold through day 29, at which point tumors from all mice in the vector control group had exceeded 1500mm 3 and were euthanized (FIG. 40B). These results indicate that in the TC-1 tumor model the Ad5 [E1-, E2b-]-E6/E7 immunotherapeutic was a potent inhibitor of tumor growth and lead to significant overall survival benefit, however complete clearance of tumors was only observed when treatment was initiated in smaller tumors. Furthermore, these results demonstrate that, despite the presence of immune suppressing PDL1 on tumor cells, immunotherapeutic treatment with Ad5 [El -, E2b-]-E6/E7 resulted in significant inhibition of tumor growth. Combination immunotherapy with immune checkpoint inhibition

[00572] To determine if the therapeutic effect of Ad5 [E1 -, E2b-]-E6/E7 could be improved in the setting of large tumors, anti-PDl antibody was co-administered. Four groups of mice (n=7/group) were implanted with 2x10 5 TC-1 tumor cells on day 0 and beginning on day 10 the mice received weekly administrations of SQ 10 10 VP Ad5 [E1-, E2b-]-E6/E7 plus IP \00μg anti-PDl, 10 10 VP Ad5 [E1-, E2b-]-null plus 10(^g anti-PDl, 10 10 VP Ad5 [E1-, E2b-]-E6/E7 plus 100 μg rat IgG 2a isotype control, or 10 10 VP Ad5 [E1-, E2b-]-null plus 100 μg rat IgG 2a isotype control. Tumor size was monitored over time and mice were euthanized when tumor size exceeded 1500 mm 3 or when tumor ulceration was present. Control mice that received Ad5 [E1 -, E2b-]-null plus 100 μg rat IgG 2a isotype control (FIG. 41A) and mice treated with Ad5 [E1-, E2b-]-null plus 100μg anti-PDl (FIG. 41B) exhibited a similar tumor growth pattern. No significant survival benefit was observed between these two groups. Mice that received Ad5 [E1 -, E2b-]-E6/E7 plus rat IgG 2a isotype control had a delayed tumor growth pattern as compared to the controls and 2 of the mice had tumor regressions to near baseline level at day 52 post tumor implantation (FIG. 41C). Four of the 7 mice that received Ad5 [E1-, E2b-]-E6/E7 and anti-PDl had tumor regression starting at day 25, and two of these resulted in tumor clearance through the end of experiment at day 53 (FIG. 41D).

[00573] Mice treated with Ad5 [E1-, E2b-]-E6/E7 plus rat IgG 2a isotype control (FIG. 42) also experienced a survival benefit with 28.6% of the animals surviving at termination of the study whereas 100% of the control mice (Ad5 [E1-, E2b-]-null plus rat IgG 2a isotype control) and the Ad5 [E1-, E2b-]-null plus anti-PDl treated mice had to be terminated by day 28 and 32, respectively (FIG. 42). Mice treated with both Ad5 [E1 -, E2b-]-E6/E7 and anti-PDl antibody had the greatest treatment benefit (FIG. 42), demonstrating delayed tumor growth and a significant improvement (P < 0.0006) in survival as compared to the controls.

[00574] Mouse anti-rat IgG antibody responses were induced by the second injection (endpoint antibody titer 1 :200 by ELISA, data not shown) with rat anti-PDl antibody and these responses were dramatically increased by the third injection (endpoint antibody titer 1 :4000 to 1 : 8000 by ELISA, data not shown). This anti-rat antibody response may explain why no anti-tumor activity was observed after injections with anti-PDl antibody alone. Also, it is likely that the first and possibly the second injections of anti-PDl antibody combined with Ad5 [E1-, E2b-]-E6/E7 immunotherapy were effective but the third injection with anti-PDl was effectively neutralized by the induced mouse anti-rat IgG response.

Tumor microenvironment following combination immunotherapy

[00575] To analyze cell populations that contributed to delayed tumor growth and survival in Ad5 [E1-, E2b-]-E6/E7 treated mice, tumor infiltrating lymphocytes (TILs) were by flow cytometry. Four groups of mice were implanted with 2x10 5 TC-1 cells and began treatment 10 days later with two weekly immunizations of Ad5 [E1-, E2b-]-E6/E7 plus PD1 antibody. On day 27 whole tumors were collected and processed as described in the materials and methods. The number of infiltrating CD8 + T cells per mg of tumor was significantly increased in the Ad5 [El -, E2b-]-E6/E7 treated groups as compared to the groups that received Ad5 [E1-, E2b-]-null (FIG. 43C). Anti-PDl antibody treatment had little or no effect on the number of infiltrating CD8 + T cells (FIG. 43C). There was no difference between any of the four groups, in terms of the number of infiltrating Tregs

(CD4 + CD25 + Foxp3 + ) per mg of tumor (FIG. 43B). However, the increase in CD8 + T cells led to a decrease in the Treg:CD8 + T cell ratio in the tumor microenvironment when the mice were treated with the Ad5 [E1-, E2b-]-E6/E7 vaccine or Ad5 [E1-, E2b-]-E6/E7 vaccine plus anti-PDl antibody treatment (FIG. 43A).

[00576] To further study the synergistic/additive effect of anti-PDl antibody to Ad5 [E1-, E2b-]- E6/E7 immunotherapy, the expression of PD1, LAG-3, and CTLA-4 was examined on TILs. The expression of these co-inhibitory molecules on T cells within the tumor microenvironment has been shown to down regulate activation of antigen-specific T cells. Immunizations with Ad5 [E1 -, E2b-]- E6/E7 plus control antibody treatment significantly increased the fraction of PD1 + and LAG-3 + CD8 + TILs, whereas, expression of these co-inhibitory molecules on CD4 + TILs was unaffected by this treatment. The percentage of CD4 + and CD8 + TILs expressing CTLA-4 was not significantly affected by vaccine treatment (data not shown). Combining anti-PDl antibody injections with Ad5 [E1-, E2b-]-E6/E7 vaccine treatment resulted in a significant reduction in the fraction of PD1 + CD8 + and CD4 + TILs, as compared with those found in tumors from mice treated with Ad5 [El -, E2b-]- E6/E7 plus control antibody (p=0.0083 for CD8 + TILs and p=0.0016 for CD4 + TILs). Furthermore the fraction of PD1 + CD8 + TILs was decreased to the level of expression observed in the Ad5 [E1-, E2b-]-null treated control groups, and the fraction of PD1 + CD4 + TILs was significantly reduced to below that observed in the control groups (p=0.0016, FIG. 44A). In addition, the percentage of LAG-3 + CD8 + TILs was also observed to decrease when the Ad5 [E1-, E2b-]-E6/E7 immunization was combined with the anti-PDl checkpoint inhibitor (p=0.0363, FIG. 44B). Since it has previously been shown that vaccine treatment can enhance PDL1 expression on tumor cells ex vivo, the expression of PDL1 was examined on tumor cells. There was an augmentation in the median fluorescence intensity of PDL1 on tumor cells after vaccine treatment. However, PDL1 expression was reduced in mice treated with the combination of Ad5 [E1-, E2b-]-E6/E7 and anti-PDl antibody, although this level was still significantly expressed above that observed in Ad5 [E1 -, E2b-]-null treated control mice.

[00577] In summary, the data demonstrate that Ad5 [E1-, E2b-]-E6/E7 can induce HPV-E6/E7 directed CMI responses in a dose dependent manner, which results in upregulation of PDL1 on tumor cells. Multiple homologous immunizations in tumor bearing mice with the highest dose of vaccine resulted in significant anti-tumor activity and increased survival, particularly in mice bearing small tumors. Importantly, a greater degree of anti-tumor activity was achieved when immunotherapy with Ad5 [E1-, E2b-]-E6/E7 was combined with anti-PDl in mice with large tumors. Overall, immunizations with the Ad5 [E1-, E2b-]-E6/E7 vaccine combined with anti- PDl antibody results in an increase in CD8 + and CD4 + effector populations that have a less exhaustive/anergic phenotype and therefore favor the balance to a more pro-inflammatory state in the tumor microenvironment. The observation that the combined treatment was associated with reductions in large tumor mass indicates that immunotherapy with Ad5 [E1-, E2b-]-E6/E7 combined with anti-PDl antibody might increase clinical effectiveness during the

immunotherapy of patients with HPV-associated head and neck or cervical cancers.

Furthermore, the data suggests that clinical trials with the Ad5 [E1-, E2b-]-E6/E7 vaccine should be combined with an immune pathway checkpoint modulator and remains a high priority.

EXAMPLE 11: Clinical Trial ofETBX-041 (Ad5 [E1-, E2b-]-HPV-E6/E7 Vaccine

[00578] The objective of the study is to evaluate the safety and immunogenicity of

immunizations with the ETBX-041 (Ad5 [E1-, E2b-]-HPV-E6/E7) vaccine in patients that are human papilloma virus type 16 (HPV-16) positive, in patients with HPV associated head and neck squamous cell carcinoma (HNSCC), and in patients with HPV associated cervical cancer. Current interventions in HNSCC patients include therapy with cisplatin and radiation. However, many HNSCC patients that initially respond or do not respond ultimately relapse. The ETBX- 041 vaccine is designed to induce anti -tumor T cell -mediated immune responses directed against the early 6 (E6) and early 7 (E7) genes of HPV. One of the important features of the ETBX-011 vaccine is that it can be combined with chemotherapy/radiation treatment.

[00579] ETBX-041 is an adenovirus serotype 5 (Ad5) vector that has been modified by removal of the El, E2b and E3 genes and insertion of a modified fused non-oncogenic HPV- E6/E7 gene. The resulting recombinant replication-defective vector can only be propagated in the newly engineered, proprietary human 293 based cell line (E.C7) that supplies the El and E2b gene functions in trans required for vector production.

[00580] A Phase I/II clinical trial of ETBX-011 (Ad5 [E1-, E2b-]-CEA) was performed, which employs the same Ad5 vector backbone that expresses a modified tumor associated antigen carcinoembryonic antigen (CEA(6D)). The maximum dose level of 5 x 10 11 viral particles (VP) was reached without any reported toxicity including dose-limiting toxicity. CEA-specific T cell responses by ELISpot have been observed in treated patients. The safety profile from the treatment of over expressing CEA cancer patients using the same Ad5 vector backbone gives us a rationale to use the same Ad5 backbone expressing HPV-E6/E7 for the treatment of patients with HNSCC cancers expressing HPV-E6/E7 to evaluate the safety and immunogenicity. No gene transfer insertion is proposed for this protocol; the product functions and remains episomal. [00581] The ETBX-041 vaccine product is used to induce HPV-E6/E7 specific cell mediated immune responses in a safe and effective manner in patients. An open-label, dose-escalation clinical study will be conducted to evaluate the safety and immunogenicity of ETBX-041 injections. The dosage levels to be evaluated are 5xl0 10 , lxlO 11 and 5xl0 u virus particles (VP) of ETBX-041. Patients will be enrolled into successive increasing dosage levels involving three (3) cohorts of patients that will be monitored for dose-limiting toxicity (DLT). Each patient will be given ETBX-041 by SQ injection every 3 weeks for 3 immunizations. Assessment of DLT for dose escalation will be made after all patients in a cohort have had a study visit at least 3 weeks after receiving their last dose of vaccine.

[00582] A total of 94 immunization treatments were administered to all patients. There was no dose-limiting toxicity and no serious adverse effects (SAE) that resulted in treatment discontinuation at any vaccine dose level. The most common toxicity was a self-limited, injection site reaction. Other reactions occurred with less than a 10% incidence and included fever, flu-like symptoms, anorexia, chills, nausea, and headache. These symptoms were also self-limiting and did not require intervention other than symptomatic measures such as acetaminophen.

[00583] The safety profile from the treatment of over expressing CEA cancer patients using the same Ad5 vector backbone gives us a rationale to use the same Ad5 backbone expressing HPV- E6/E7 for the immunization (vaccination) of patients that are HPV- 16+ and at high risk for developing HPV+ cancers or who have HPV+ cancers.

[00584] Preclinical studies

[00585] Studies were performed to assess the use of Ad5 [E1-, E2b-]-HPV-E6/E7 as a cancer vaccine in a C57B1/6 mouse model. Ad5 [E1-, E2b-]-HPV-E6/E7 induced potent CMI against HPV-E6/E7 in mice. The Ad5 [E1-, E2b-]-HPV-E6/E7 significantly inhibited progression of established HPV-E6/E7 tumors in a murine model of HPV E6/E7 expressing cancer. These data indicate that in vivo delivery of Ad5 [E1-, E2b-]-HPV-E6/E7 can induce anti-HPV E6/E7 immunity and inhibit progression of HPV E6/E7 expressing cancers.

[00586] Dose Response of Ad5 [E1-, E2b-]-HPV-E6/E7 A study was performed to determine the effect of immunizations with increasing doses of Ad5 [E1-, E2b-]-HPV-E6/E7 on the induction of cell- mediated immune (CMI) responses in Ad5 naive mice. Groups of Ad5 naive BALB/c mice (n=5/group) were immunized subcutaneously (SQ) three times at 2-week intervals with 10 8 , 10 9 , or 10 10 VP of Ad5 [E1-, E2b-]-HPV-E6/E7, respectively. Control mice were injected with buffer solution only. Two weeks after the last immunization, T cell CMI responses were assessed by IFNy ELISpot analysis. As shown in FIG. 36, a dose response effect was observed on the CMI response with the highest CMI response level being obtained after immunizations with 10 10 VP using Ad5 [E1-, E2b-]-HPV-E6/E7 immunization. Specificity studies revealed that CMI responses were specific to HPV E6 and E7 and there were no responses against the irrelevant antigen SIV-1 nef.

[00587] Induction of CMI responses after Ad5 [El-, E2b-]-HPV-E6/E7 vaccination as assessed by flow cytometry. To assess CMI induction by flow cytometry following multiple homologous

immunizations with Ad5 [E1-, E2b-]-HPV-E6/E7, groups of C57B1/6 mice (n=5/group) were immunized three times SQ at 2-week intervals with 10 10 VP of Ad5 [E1-, E2b-]-HPV-E6/E7. Two weeks following the last immunization, splenocytes were exposed to HPV-E6/E7 peptides or irrelevant antigens and analyzed by flow cytometry for the number of IFN-γ and/or TNFa expressing T cells. As shown in FIG. 68, both IFN-γ and/or TNFa expressing T cells were induced as a result of multiple homologous immunizations with the highest dose of Ad5 [E1-, E2b-]-HPV-E6/E7. Specificity studies revealed that CMI responses were specific to HPV E6 and E7 and there were no responses against irrelevant antigens such as SIV-vif or SIV-nef.

[00588] Toxicology. An extensive pre-clinical toxicology study will be conducted to assess the toxicity of Ad5 [E1-, E2b-]-HPV-E6/E7 following SQ injections on in C57/B1/6 mice. Toxicity endpoints will be assessed at various time points post-injection. The animals will be

administered up to 3 SQ injections on Days 1, 22 and 43, with either vehicle control or Ad5 [El- , E2b-]-HPV-E6/E7 at a dose consistent with that to be used in clinical trials accounting for difference in body mass. Evaluations will consist of effects on body weights, body weight gain, food consumption pathology, blood hematology analyses, blood chemistry analyses, and test on coagulation time.

[00589] Treatment of established HPV E6/E 7 expressing tumors with vaccine alone

[00590] The effectiveness of treating established HPV E6/E7 expressing tumors in vivo with Ad5 [E1-, E2b-]-HPV-E6/E7 was evaluated. C57B1/6 mice were implanted SQ into the right subcostal with 10 6 HPV E6/E7 expressing tumor cells on day 0. Tumors were palpable by days 4-6. On days 6, 13, and 20, mice were treated by SQ injections of 10 10 VP of Ad5 [E1-, E2b-]- null (empty vector controls) or 10 10 VP of Ad5 [E1-, E2b-]-HPV-E6/E7. All mice were monitored for tumor growth and tumor volumes were calculated. As shown in FIG. 69, mice immunized with Ad5 [E1-, E2b-]-HPV-E6/E7 had significantly smaller tumors than control mice (p<0.01). These results demonstrate that the Ad5 [E1-, E2b-]-HPV-E6/E7 vector platform has the potential to be utilized as an immunotherapeutic agent to treat HPV-E6/E7 expressing tumors.

[00591] Treatment of established HPV E6/E 7 expressing tumors with vaccine and

chemotherapy/radiation treatment. The effectiveness of treating HPV E6/E7 expressing tumors in vivo with Ad5 [E1-, E2b-]-HPV-E6/E7 combined with chemotherapy/radiation treatment was evaluated. C57B1/6 mice were implanted SQ with 10 6 HPV E6/E7 expressing tumor cells on day

0. Established HPV-E6/E7 expressing tumors were treated by immunotherapy on days 7, 14, and 21 combined with cisplatin/radiation treatment on days 13, 20, and 27. Control tumor bearing mice were treated by injections with Ad-null combined with cisplatin/radiation treatment. As shown in FIG. 52, combination treatment using Ad5 [E1-, E2b-]-HPV-E6/E7 and chemotherapy/radiation resulted in significant extension of survival time as compared with control mice receiving treatment Ad5 [E1-, E2b-]-null (Empty vector control) and

chemotherapy/radiation. These results show that vaccine immunotherapy can be combined with chemotherapy/radiation treatment and that this combination results in a significantly greater extension of survival in a mouse model of HPV E6/E7 expressing cancer.

[00592] In summary, ETBX-041 (Ad5 [E1-, E2b-]-HPV-E6/E7) is a non-oncogenic vaccine targeting HPV-E6 and E7 that induces robust immune responses. Ad5 [E1-, E2b-]-HPV-E6/E7 induced potent CMI against HPV-E6/E7 in mice assessed in ELISpot and flow cytometry studies. Ad5 [E1-, E2b-]-HPV-E6/E7 significantly inhibited progression of established tumors in a murine model of HPV E6/E7 expressing cancer. Immunotherapy with Ad5 [E1-, E2b-]-HPV- E6/E7 could be combined with chemotherapy/radiation treatment to significantly increase survival in tumor bearing mice. The goal is to further develop this novel Ad5 vector system that overcomes barriers found with other Ad5 systems and clinically tests this vaccine to determine that significant HPV-E6/E7 directed immune responses are induced in immunized (vaccinated) individuals. The results of this clinical study will establish the safety and immunogenicity of using this new Ad5 [E1-, E2b-]-HPV-E6/E7 vaccine.

EXAMPLE 12: Preclinical data of Ad-CEA Vaccine

[00593] Ad5 Vaccine Expressing CEA (ETBX-011) : The Ad5 [E1-, E2b-]-CEA(6D) vaccine

(ETBX-011) is an adenovirus vector vaccine in which the El, E2b and E3 gene regions have been removed and replaced with a gene encoding CEA with the CAP1-6D mutation.

[00594] ETBX-011 vaccine is supplied as a sterile, clear solution in a single-dose vial. Vials will contain a single dose of 5x1011 VP. Dose will be administered according to the clinical protocol.

The maximum tolerated dose will be determined in a dose escalation study as outlined in the clinical protocol.

[00595] Instructions for Storage, Handling and Administration

[00596] ETBX-011 vaccine should be stored at -20 °C.

[00597] Prior to injection, the appropriate vial should be removed from the freezer and allowed to thaw at controlled room temperature (20-25 °C, 68-77 °F) for at least 20 minutes and not more than

30 minutes, after which it should be kept at 28 °C (35 46 °F). The vaccine is stable for at least 8 hours after removal from the freezer when kept refrigerated at 28 °C (35-46 °F).

[00598] The thawed vial should be swirled and then, using aseptic technique, the pharmacist should withdraw the appropriate volume (1.0 mL) from the vial using a 1 mL syringe. The vaccine should be injected as soon as possible using a 1 to 1/2 inch, 20 to 25 gauge needle. If the vaccine cannot be injected immediately, the syringe should be stored at 28°C (35-46°F).

[00599] All injections of vaccine should be given as a volume of 1.0 mL by subcutaneous injection in the upper arm after preparation of the site with alcohol. Either arm may be used for each injection.

[00600] When preparing a dose in a syringe and administering the dose, consideration should be given to the volume of solution that may remain in the needle after the dose is administered, to ensure that the full dose specified in the protocol is administered.

[00601] Immunization with Ad5-CEA(6D) platforms: In pre-clinical studies, the

immunogenicity of Ad5 [E1-, E2b-]-CEA(6D) vaccine was tested in an Ad5 naive and an Ad5 immune murine model. To assess the induction of cellular meditated immunity following immunization, Ad5 naive C57B1/6 mice (n=7/group) were immunized three times

subcutaneously (SQ) with 10 10 VP of Ad5 [El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D), or injection buffer alone (control) at weekly intervals. Three immunizations were used because prior studies in an HIV-1 model demonstrated that the Ad5 [E1-, E2b-] vector induced a maximum immune boost after three immunizations. Fourteen days after the final immunization, CEA-specific CMI responses were determined by IFN-γ and IL-2 ELISpot assays. Mice immunized with Ad5 [E1-, E2b-]-CEA(6D) induced a significantly greater number of IFN- γ (p<0.01) and IL-2 (p<0.01) secreting splenocytes than mice immunized with Ad5 [El -]- CEA(6D) (FIG. 45A and FIG. 45B). Splenocytes from vaccinated mice and controls were assessed for non-specific cytokine secretion following stimulation with HIV-1 Gag and β- galactosidase. Non-specific IFN-γ or IL-2 secretion was not detected in T-cells from the spleens of mice vaccinated with either CEA expressing vector (FIG. 45A and FIG. 45B). Splenocytes were also stimulated with Ad5-null to confirm positive vaccination against Ad5 and splenocytes derived from all such immunized mice harbored Ad5 specific T-cells (FIG. 45A and FIG. 45B). Elevated levels of CEA specific IgG antibody were detected after immunizations and these levels were comparable for both Ad5-CEA vectors.

[00602] CMI against Ad5 [El-]-CEA(6D) andAdS [E1-, E2b]-CEA(6D) in Ad5 immune mice: To assess whether Ad5 [E1-, E2b-]-CEA(6D) is more effective than Ad5 [El-]-CEA(6D) in the presence of Ad5 immunity, Ad5 naive C57B1/6 mice (n=7/group) were immunized twice with 10 10 VP of Ad5 [El-]-null to induce Ad5 immunity. The presence of anti-Ad5 IgG antibody was confirmed by ELISA as 0.028 ± 0.028 ng Equivalents of IgG antibody for baseline samples versus 39.50 ± 3.78 ng Equivalents of IgG antibody in samples after Ad5-null immunizations (p<0.0001). Ad5 immune mice were then immunized three times at weekly intervals with 10 10 VP of Ad5 [El-]-CEA(6D) or Ad5 [E1-, E2b-]-CEA(6D). Controls received injection buffer alone. Splenocytes were collected 14 days after the final immunization and assessed by IFN-γ and IL-2 ELISpot. Ad5-immune mice immunized with Ad5 [E1-, E2b-]-CEA(6D) exhibited significantly higher levels of IFN-γ (p=0.04) and IL-2 (p<0.01) secreting splenocytes as compared to Ad5 immune mice immunized with Ad5 [El-]-CEA(6D) (FIG. 46A and FIG. 46B). Splenocytes were also assessed for non-specific secreting T-cells of IFN-γ (FIG. 46A) or IL-2 (FIG. 46B) by stimulation with the non-immunizing antigens Beta-galactosidase (β-gal) and HIV-1 Gag. When compared to the results for naive mice above, levels of CEA specific IgG antibody generated after immunizations were greatly reduced.

[00603] Ability of the Ad5 [E1-, E2b-] -CEA(6D) vector to break tolerance: Having

immunologically compared the two Ad5-CEA(6D) vectors, a study was then performed to assess if the Ad5 [E1-, E2b-]-CEA(6D) vector could break tolerance in the more stringent CEA transgenic mouse model in which CEA is a self-antigen. Mice were immunized once with 2.6 X 10 10 VP of Ad5 [E1-, E2b-]-CEA(6D) and 14 days later, their splenocytes were assessed for CMI responses by IFN-γ ELISpot assays. Elevated numbers of IFN-γ secreting splenocytes were observed after only one injection with the vector platform (FIG. 47). ELISpot studies employing Cytomegalovirus (CMVpp65) and HIV-gag antigens showed that the response was specific to CEA.

[00604] Treatment of CEA expressing tumors in Ad5 naive mice: The efficacy of

immunizations with Ad5 [E1-, E2b-]-CEA(6D) was evaluated by the treatment of CEA expressing MC-38 tumors that were growing in Ad5 naive and Ad5 immune mice. A group of C57B1/6 mice (n=7) were implanted SQ in the flank with 10 6 CEA expressing MC-38 tumor cells (MC38-CEA). Six days later, when tumors were palpable (3-5mm), the mice were treated with three SQ injections of 10 10 VP of Ad5 [E1-, E2b-]-CEA(6D) on days 7, 13 and 19.

Untreated C57B1/6 mice (n=7) implanted with 10 6 MC38-CEA cells and injected at the same time points with injection buffer alone served as controls. Tumor volumes were measured and reported as previously described. Briefly, tumors were measured by two opposing diameters and reported as a multiple of those numbers to determine tumor volumes. Statistical differences were determined using the Bonferroni posttests analysis with PRISM software. The sizes of treated and control tumors are shown in FIG. 48A as a function of time. At Day 20 post implant the tumors in mice treated with Ad5 [E1-, E2b-]-CEA(6D) were significantly (p<0.05) smaller as compared to controls (FIG. 48A). Tumors were excised and weighed at the termination of the study. Tumors from mice treated with Ad5 [E1-, E2b-]-CEA(6D) weighed significantly (p<0.05) less than tumors from control mice (FIG. 48B). Splenocytes from both groups of mice were assessed for the number of CEA stimulated IFN-γ secreting T cells by ELISpot. Mice immunized with Ad5 [E1-, E2b-]-CEA(6D) had a significantly (p<0.05) greater number of INF - γ secreting T-cells in their spleens as compared to control mice (FIG. 48C).

[00605] Treatment of CEA expressing tumors in Ad5 immune mice: To determine if Ad5 immune MC38-CEA tumor bearing mice could be treated with Ad5 [E1-, E2b-]-CEA(6D), C57B1/6 mice (n=7) were immunized two times at a two-week interval with 10 10 VP of Ad5 [El-]-null. Two weeks after the second Ad5 [El-]-null immunization, mice were implanted SQ with 10 6 CEA expressing MC-38 tumor cells. Six days later tumors were measurable (3 -5mm) and the mice were treated with three SQ injections of 10 10 VP of Ad5 [E1-, E2b-]-CEA(6D) at weekly intervals. C57B1/6 mice (n=7) which were immunized twice with Ad5 [El-]-null, inoculated with 10 6 MC38-CEA cells and injected three times SQ with injection buffer alone served as controls. All mice were monitored for tumor growth and tumor volumes are reported using previously described methods. Briefly, tumors were measured by two opposing diameters and reported as a multiple of those numbers to determine tumor volumes. Statistical differences were determined using the Bonferroni posttests analysis with PRISM software. Evaluation was over a 21 -day period and the size of MC38-CEA tumors was determined as a function of time. At day 19 the tumors in mice treated with Ad5 [E1-, E2b-]-CEA(6D) were significantly smaller as compared to the controls (FIG. 49A). At the end of the study all mice were sacrificed and the tumors were excised and weighed. Tumors from the mice treated with Ad5 [E1-, E2b-]- CEA(6D) weighed significantly less than the tumors from controls (FIG. 49B). The number of CEA stimulated T-cells in the spleen which secreted IFN-γ was determined by ELISpot assay. Mice immunized with Ad5 [E1-, E2b-]-CEA(6D) had a significantly greater number of IFN-γ secreting T-cells than did control mice that received injection buffer alone (FIG. 49C).

[00606] In summary, Ad5 [E1-, E2b-]-CEA(6D) is a therapeutic vaccine targeting CEA that induces robust immune responses, even in the presence of pre-existing Ad5 immunity. When compared directly with a conventional Ad5 [El-]-CEA(6D) vector, the Ad5 [E1-, E2b-]- CEA(6D) platform induced superior cellular mediated immune responses in both Ad5 naive and Ad5 immune animals. In therapeutic efficacy studies, treatment with Ad5 [E1-, E2b-]-CEA(6D) resulted in significantly decreased established MC38-CEA tumor growth even in mice which had been pre-immunized against Ad5. Treatment with Ad5 [E1-, E2b-]-CEA(6D) did not result in any adverse effects in pre-clinical studies. The results of these pre-clinical studies exemplify the technical and immunological merit of using the Ad5 [E1-, E2b-]-CEA(6D) vaccine as a therapeutic agent. EXAMPLE 13: Clinical study of Ad-CEA(6D) Vaccine

[00607] Clinical safety : Phase I/II clinical trial of Ad-CEA (Ad5 [E1-, E2b-]-CEA(6D)) (IND#14325) have been performed. The Phase VII study consisted of a dose-escalation study of four dosage levels (lxlO 9 , lxlO 10 , lxlO 11 , 5xl0 u VP/dose) of ETBX-011 (Phase I component), and the maximally tolerated dose of ETBX-011 (Phase II and 5x10 11 VP/dose components). Ad- CEA was administered by SQ injection every 3 weeks. Thirty-two patients with metastatic colorectal cancer, median age 57.5 (range 38-77) who had failed a median of three prior chemotherapeutic regimens (range 2-5), had a performance status of 90 % (range 70-100 %), and had three sites of metastatic disease (range 1-4), were enrolled. The majority of patients were able to receive all three immunizations. Four patients who stopped immunizations early did so due to significant disease progression.

[00608] A total of 94 immunization treatments were administered to all patients. There was no dose-limiting toxicity and no serious adverse effects (SAE) that resulted in treatment discontinuation at any vaccine dose level. The most common toxicity was a self-limited, injection site reaction. Other reactions occurred with less than a 10% incidence of all adverse effects (AE) reported and included fever, flu-like symptoms, anorexia, chills, nausea, and headache. These symptoms were also self-limiting and did not require intervention other than symptomatic measures such as acetaminophen.

[00609] CMI response in treated patients: CEA specific immune responses following immunization treatments with the product candidate were evaluated. As determined by an ELISA technique, no antibody activity directed against CEA was observed. CMI responses in colorectal cancer patients treated in cohort 1, cohort 2, cohort 3/Phase II, and cohort 5 was assessed. PBMCs were isolated prior to immunotherapy treatment and after all treatments as well as three weeks following the last treatment from patients. CEA specific ELISpot assays were performed on PBMC to determine the numbers of interferon gamma (IFN-γ) secreting lymphocytes (SFC) after exposure to CEA peptides in vitro. The highest CMI responses during immunizations, regardless of time point (weeks 3, 6, or 9) in the patients treated in cohort 1, cohort 2, cohort 3/Phase II, and cohort 5 was determined. This analysis revealed a dose response to increasing levels of product (FIG. 14). The highest CMI levels occurred in patients that received the highest dose of 5xl0 u VP (Cohort 5). In a preliminary study, it was observed that a population of polyfunctional CD8+ T cells (those that secrete more than 1 cytokine when activated) after immunizations secreted multiple cytokines, a sign of greater functionality of T cells induced by the vaccine. In further follow-up analysis of a few patient blood samples, a decrease in CEA directed after immunotherapy was stopped was noted (FIG. 50). This observation supports a rationale for booster immunizations. [00610] Ad5 NAb (neutralizing antibodies) and CMI against Ad5 were also measured and correlated with CEA-specific CMI. Each patient had their serum and PBMC sample tested at baseline (prior to treatment) and at 9 weeks after completion of 3 treatments. Nineteen of 31 colorectal cancer patients (61%) tested in this study had Ad5 neutralizing activity in serum samples prior to the onset of treatment with Ad5 [E1-, E2b-]-CEA(6D). The mean pre-treatment Ad5 NAb titer value obtained among all patients was 1 : 189 ±1 :71 SEM and the mean pre- treatment Ad5 NAb titer among seropositive patients was 1 :308 ± 1 : 108. Analysis of serum samples from patients who received 3 immunizations revealed Ad5 NAb titers that were significantly increased (PO.0001, Mann-Whitney test) by week 9 (mean 1 :4767 ± 1 : 1225 SEM) when compared with their respective baseline values. Analysis of PBMC for CMI responses to Ad5 also revealed a significant increase (P<0.01, Mann-Whitney test) in Ad5 directed CMI responses after immunizations with Ad5 [E1-, E2b-]-CEA(6D) (22.6 ± 9.3 SEM IFN-γ secreting SFC at week 0 versus 191.1 ± 83.7 IFN-γ SFC at week 9).

[00611] Clinical evidence of activity: The Ad-CEA treated colorectal cancer patients (total=32) were followed for survival and Kaplan-Meier plots and survival proportions performed (PRISM software). Events were determined by information from the social security death index (SSDI) database, clinical charts and telephone calls (FIG 51A-FIG. 51C).

[00612] The seven patients in cohorts 1 and 2 experienced a 12-month survival proportion of 29%. The 21 patients in cohort 3 and Phase II experienced a 12-month survival proportion of 48%. The six patients in cohort 5 experienced a 12-month survival proportion of 50%. Twenty-nine month overall survival of the intent-to-treat population (32 patients) was 20% (FIG. 51 A) with a median survival time of 11 months from informed consent/first injection. For the subset of 28 patients that received all 3 immunizations, the 29-month survival was 23% (FIG. 51B) with a median survival time of 13 months. For the 22 patients optimally dosed with the two highest doses of vaccine (1 and 5 x 10 11 ) and receiving all 3 immunizations, the 28-month overall survival was 19%) (FIG. 51C). Median overall survival was 13 months in the optimally treated patients. There were 3 stable disease events observed immediately after completion of treatment.

[00613] Evidence of an Ad5 [E1-, E2b-J -based vaccine driving T cell infiltration and PD-L1 upregulation: High-risk human papillomavirus (HPV) such as HPV type- 16 is associated with the etiology of cervical and more than 90% of HPV-related head and neck squamous cell carcinomas (HNSCC). The HPV early 6 (E6) and early 7 (E7) genes are expressed at high levels in HPV- induced cancers and are involved in the immortalization of primary human epidermal cells. The Ad5 [E1-, E2b-]-based vector platform was investigated in a murine model of HPV-associated E6/E7 expressing cancer. The immunotherapy against human papilloma virus (HPV) using a viral gene delivery platform to immunize against HPV 16 genes E6 and E7 (Ad5 [E1-, E2b-]-E6/E7) combined with programmed death-ligand 1 (PD-1) blockade increased therapeutic effect as compared to the vaccine alone. Ad5 [E1-, E2b-]-E6/E7 as a single agent induced HFV-E6/E7 cell-mediated immunity. Immunotherapy using Ad5 [E1-, E2b-]-E6/E7 resulted in clearance of small tumors and an overall survival benefit in mice with larger established tumors. When immunotherapy was combined with immune checkpoint blockade, an increased level of anti-tumor activity against large tumors was observed (FIG. 44A- FIG. 44C). Analysis of the tumor microenvironment in Ad5 [E1 -, E2b-]-E6/E7 treated mice revealed elevated CD8 + tumor infiltrating lymphocytes (TILs); however, induction of suppressive mechanisms such as programmed death-ligand 1 (PD-L1) expression was observed on tumor cells and an increase in PD-1 + TILs was also shown. When Ad5 [E1-, E2b-]- E6/E7 immunotherapy was combined with anti-PD-1 antibody, it was observed that CD8 + TILs at the same level but a reduction in tumor PD-L1 expression on tumor cells and reduced PD-1 + TILs providing a mechanism by which combination therapy favors a tumor clearance state and a rationale for pairing antigen-specific vaccines with checkpoint inhibitors in future clinical trials.

[00614] Combining chemotherapy treatment with Ad5 [E1-, E2b-]-based vaccine immunotherapy: Using the same murine model of HPV-associated E6/E7 expressing cancer described above, immunotherapy using Ad5 [E1-, E2b-]-E6/E7 combined with cisplatin/radiation treatment that is utilized to treat HNSCC was investigated. When immunotherapy was combined with

cisplatin/radiation treatment, a significant increase in survival time demonstrated in tumor bearing mice treated with the combination of immunotherapy and cisplatin/radiation treatment as compared to control mice receiving cisplatin/radiation treatment alone was seen (FIG. 52).

[00615] The effects of combining immunizations with Ad5 [E1-, E2b-]-E6/E7 with

cisplatin/radiation treatment versus cisplatin/radiation treatment alone in a murine model was assessed. The combination of Ad5 [E1-, E2b-]-E6/E7 immunizations plus cisplatin/radiation treatment resulted in the induction greater CMI responses as compared to immunizations with Ad5 [E1-, E2b-]-E6/E7 alone (FIG. 53). These results provide a rationale for combining immunotherapy with chemotherapy treatment in order to achieve greater anti-tumor CMI responses.

[00616] Toxicology

[00617] Because of the possibility that the lower host reactivity against the Ad5 [E1-, E2b-]-

CEA(6D) could result in an altered hepatic toxicity profile, hepatic adverse effects were determined following immunization with the CEA expressing vectors or injection buffer alone in Ad5-na ' ive and Ad5-immune mice. Studies to evaluate adverse hepatic effects of vaccination were performed first using Ad5-naive mice immunized at weekly intervals with 1010 VP of Ad5

[El-]-CEA(6D), Ad5 [E1-, E2b-]-CEA(6D) or controls injected with injection buffer alone.

Three days after one immunization (Day 3) or three immunizations (Day 17), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were assessed in blood samples to determine liver adverse effects due to treatment. All liver enzyme levels were within normal range after immunization with either vector platform (FIGS. 64A-64B). Liver necrosis is determined by a change in the AST/ ALT ratio from greater than one to less than one. There was no indication of liver necrosis following vector immunization or in control animals.

[00618] Studies of hepatic adverse effects were also performed using Ad5 immune mice.

C57B1/6 mice (n=7) were injected twice at a two week interval with 1010 VP of Ad5 [El -]-null and then immunized three times with Ad5 [El-]-CEA(6D) or Ad5 [E1-, E2b-]-CEA(6D) as described above. A group (n=7) of Ad5 naive and Ad5 immune mice immunized with injection buffer alone served as controls. Three days after the last immunization, ALT and AST levels were assessed on the blood samples to determine hepatic adverse effects due to treatment

(FIGS. 65A-65B). AST and ALT levels remained within the normal control range after immunizations with either vector. There was no change in the AST/ ALT ratios, indicating lack of liver necrosis following vector immunization in Ad5 immune animals. No hepatic adverse effects were detected following immunization by either platform as determined by little change in serum ALT and AST levels.

[00619] Clinical Trial for ETBX-011 (Ad5 [E1-, E2b-]-CEA(6D)) Vaccine

[00620] A Phase I/II clinical trial of ETBX-011 (Ad5 [E1-, E2b-]-CEA(6D)) (IND#14325) that expresses the tumor-associated antigen carcinoembryonic antigen (CEA) was performed. The Phase I/II study consisted of a dose-escalation study of four dosage levels (lxlO 9 , lxlO 10 , lxlO 11 , 5xl0 u VP/dose) of ETBX-011 (Phase I component), and the maximally tolerated dose of ETBX-011 (Phase II and 5xl0 u VP/dose components). ETBX-011 was administered by subcutaneous injection every 3 weeks for three treatments. Thirty-two patients with metastatic colorectal cancer, median age 57.5 (range 38-77) who had failed a median of three prior chemotherapeutic regimens (range 2-5), had a performance status of 90 % (range 70-100 %), and had three sites of metastatic disease (range 1-4), were enrolled. The majority of patients were able to receive all three immunizations. Four patients who stopped immunizations early did so due to significant disease progression. A total of 94 immunization treatments were administered to all patients. There was no dose-limiting toxicity and no serious adverse effects (SAE) that resulted in treatment discontinuation at any vaccine dose level. The most common toxicity was a self-limited, injection site reaction. Other reactions occurred with less than a 10% incidence and included fever, flu-like symptoms, anorexia, chills, nausea, and headache. These symptoms were also self-limiting and did not require intervention other than symptomatic measures such as acetaminophen.

[00621] The safety profile from the treatment of over expressing CEA cancer patients using the Ad5 [E1-, E2b-]-CEA(6D) vector gives us a rationale to use the vaccine for the treatment of additional patients with carcinomas expressing CEA to evaluate the safety and immunogenicity. ETBX-011 is a clear colorless liquid filled in a 2-mL amber vial containing 1.0 mL of extractable vaccine. There are total of 5xl0 u total virus particles in 1.0 mL of the product. Each vial is sealed with a rubber stopper and has a white flip off seal. End user of the product will need to flip the white plastic portion of the cap up/off with their thumb to expose the rubber stopper, and then puncture the stopper with an injection needle to withdraw the liquid. The rubber stopper is secured to the vial with an aluminum crimped seal.

EXAMPLE 14: Clinical Trial for Combination of FOLFOX-A, Ad-CEA vaccine, and Aveluntab

[00622] Objectives:

[00623] To determine if there is an improvement in time to progression among patients with metastatic colorectal cancer who are treated with standard of care + anti-PDLl monoclonal antibody + Ad-CEA therapeutic cancer vaccine compared with standard of care alone

[00624] Primary Objective:

[00625] To determine if there is an improvement in time to progression among patients with metastatic colorectal cancer who are treated with standard of care + Ad-CEA vaccine +

Avelumab compared with standard of care alone

[00626] Secondary Objective(s):

[00627] Safety of the combination versus standard of care alone

[00628] Immunologic effects of the combination compared with standard of care

[00629] Peripheral

[00630] Quantitate T cell responses against CEA, and other tumor associated antigens using methods such as intracellular cytokine staining, ELIspot, and/or others

[00631] Flow-based assay for analysis of PBMC subsets

[00632] T-cell clonal expansion assay

[00633] Serum cytokine and soluble factor analysis

[00634] ELISA for antibody generation against CEA

[00635] Tumoral

[00636] Immunohistochemistry analysis of T cell infiltrate (may include CD3, CD4, CD8, FoxP3 and others), immune regulatory markers (may include IDO, LAG3, TIM3, PD-1, PD-L1 and others)

[00637] RNA and proteomic analysis for immune signature changes

[00638] Whole genome sequencing for identification of mutation correlate with clinical outcome

[00639] Correlative analysis of immune endpoints with clinical outcomes [00640] Overall response rate (CR+PR by RECIST 1.1)

[00641] Overall survival

[00642] Eligibility:

[00643] Subjects age 18 and older with previously untreated pathologically confirmed metastatic colorectal cancer; prior adjuvant therapy is acceptable

[00644] ECOG performance status <_1

[00645] Normal organ and bone marrow function

[00646] Subjects with active autoimmune diseases requiring treatment and subjects requiring system steroids (except for physiologic doses for steroid replacement) are not allowed

[00647] Design:

[00648] This is a randomized, multicenter phase 2.5 clinical trial designed to evaluate the potential improvement in Progression Free Survival (PFS) when Avelumab and Ad-CEA vaccine are used in combination with standard of care therapy in metastatic colorectal cancer when compared with standard of care alone (FOLFOX-B).

[00649] A lead in cohort, comprising the first 6 evaluable subjects enrolled, will be treated with avelumab + Ad-CEA vaccine + standard of care in order to assess the safety of the combination.

[00650] If no more than 1 subject in the lead in cohort experiences a dose limiting toxicity attributable to the IND agents, 70 evaluable subjects will be randomized on a 1 : 1 basis to receive either Avelumab + Ad-CEA vaccine + standard of care (Arm B) or standard of care alone (Arm A)

[00651] Standard of care therapy consists of 6 - 12 two week cycles of bevacizumab +

FOLFOX (5-FU, leucovorin, oxaliplatin) followed by two week cycles of bevacizumab + capecitabine until disease progression

[00652] Subjects assigned Arm A that have progressive disease will be offered Avelumab +

Ad-CEA vaccine in combination with a standard chemotherapy regimen

[00653] Kaplan-Meier curves and a two-tailed log-rank test will be the primary analysis methods.

[00654] The accrual ceiling for the study is set at 81

[00655] Preclinical data support: Preclinical model evaluation of the combination of FOLFOX with anti-PD-Ll was performed by EMD-Serono.. The study was conducted in compliance with GLP and the global policy "Quality management system (QMS) for Merck Serono research (MSR)" of Merck KGaA.

[00656] The study investigated the potential of combining avelumab with the core components of FOLFOX (5-fluorouracil and oxaliplatin) in two separate studies (Til 0-079 & Til 1-001) conducted in mice bearing subcutaneous MC38 tumors. [00657] The therapeutic combination of oxaliplatin/5-FU and anti-PD-Ll blockade using avelumab in MC38 tumor-bearing mice yielded superior tumor growth inhibition relative to control mice and either form of monotherapy (FIG. 56). Additionally, the avelumab and oxaliplatin/5-FU

combination significantly extended survival compared to mice receiving either vehicle control or monotherapy (FIG. 57).

[00658] In both in vivo studies (TI10-079 and Til 1-001), a subset of mice in each treatment group was sacrificed to evaluate trends in immune phenotype in the spleen and tumor tissues. Using a pentamer analysis of splenic tissue, the avelumab and oxaliplatin/5-FU combination was shown to significantly elevate the precursor frequency of tumor-reactive P15E-specific CD8+ T cells (P15E is a known tumor antigen expressed in MC38 colorectal cancer tumor lines (FIG. 58). Additionally, avelumab therapy was shown to consistently and significantly increase the percentage of splenic CD8+ T cells expressing the PD-1 receptor (FIG. 59).

[00659] Consistent with the findings in the spleen, intratumoral CD8+ T cells and K1.1+ cells were elevated in mice receiving combination therapy with avelumab and oxaliplatin/5-FU (FIG. 60 and FIG. 61, respectively). Taken together, the phenotypic data from mice treated with the avelumab and oxaliplatin/5-FU combination suggest that this immunotherapeutic regimen is successfully activating immune effector cell populations with known tumoricidal potential (NKl . l+ and CD8+ T cells).

[00660] The frequency of IFN-γ producing CD8+ T effector cells directed against the pl5E tumor antigen was also measured using an ELISPOT assay that detects IFN-γ secretion as a marker of T cell activation. Using the ELISPOT assay, a strong pl 5E-specific response was detected in all treated groups in study Til 0-079 (FIG. 62). In study Til 1-001, the highest pl5E- specific response was measured in the combination treatment group (FIG. 61).

[00661] One interpretation of this data is that chemotherapy initiates a cascading sequence in which tumor cell destruction and the shedding of tumor antigen in the tumor microenvironment activates innate and adaptive immune responses to the tumor. In this scenario, PD-L1 blockade is thought to potentiate the adaptive immune response, which may indirectly activate innate effector cells such as K cells.

[00662] By blocking negative costimulation of tumor-reactive T cells mediated by the PD- 1/PD-Ll interaction, avelumab may be employed in a variety of settings and indications to maximize the potential efficacy of standard of care cytotoxic therapies as well as

immunotherapy combinations.

[00663] The use of standard chemotherapy in patients with previously untreated metastatic colorectal cancer was studied, and no difference was found in the number of 40+ different immune cell subsets pre- versus post-treatment with standard therapy (5-FU based chemotherapy). Thus, chemotherapy should not diminish the potential immune benefits of anti- PD-L1 therapy.

[00664] In summary, Avelumab is a monoclonal antibody with excellent binding capability against PD-Ll . A recently completed dose escalation of study of avelumab demonstrated safety and binding capability against PD-Ll in humans. FOLFOX + Bevacizumab is the standard of care for 1 st line metastatic colorectal cancer treatment. A preclinical model has demonstrated increased immune reactivity, decreased tumor growth rate, and improved survival in mice bearing MC38 tumors. Clinical data from patients pre- and post-treatment with FOLFOX + Bevacizumab demonstrates no significant difference in 40 immune cell subsets indicating standard agents will not negatively affect peripheral immune subsets. 5-FU and Oxaliplatin are involved in improving immune response to tumors through immunogenic cell death and immunogenic modulation. Standard of care with or without a novel immunotherapy may allow identification of longer term benefit where radiographic tumor responses are less common. Randomization Procedures

[00665] Randomization begins after the first 6 evaluable subjects are enrolled (lead in). These subjects are assigned to the Arm B regimen. Thereafter, subjects are randomized on a 1 : 1 basis to the following arms (as exemplified in FIG. 66):

[00666] Arm A: Subjects receive FOLFOX6 + bevacizumab for up to 12 2-week cycles followed by maintenance therapy with bevacizumab + capecitabine until disease progression.

[00667] Arm B: Subjects receive FOLFOX6 + bevacizumab + avelumab for up to 12 2-week cycles followed by maintenance therapy with bevacizumab + capecitabine + avelumab until disease progression.

Study Implementation

[00668] Study design: This is an open-label, randomized, multicenter phase 2.5 clinical trial designed to evaluate the potential improvement in time to progression (TTP) when avelumab is used on combination with standard of care therapy in metastatic colorectal cancer when compared with standard of care alone. Patients in the standard of care alone arm are offered the opportunity to cross-over and receive avelumab in combination with any combination of the Arm A standard of care deemed appropriate by the investigator at the time of disease progression (as defined by RECIST 1.1.). Cross-over is not offered in the case of rapid or symptomatic progression due to the need for implementation of a standard option with known efficacy. Other clinical and immunologic endpoints are captured with the goal of establishing correlative data that can later be prospectively tested in a larger phase 3 study.

[00669] A dosing regimen is exemplified in FIG. 67. [00670] Initially, 6 patients assigned to receive the combination of chemotherapy and avelumab are enrolled to evaluate safety and feasibility of this treatment schedule.

[00671] Dose Limiting Toxicity (Lead-in Period Only: The DLT evaluation period is the period from the first dose of avelumab until 4 weeks (28 days) have passed. DLT is assessed in the first 6 patients enrolled. If > 1/6 patients experience a DLT, with the combination. Subjects who do not complete the DLT observation period for reasons other than a DLT are replaced.

[00672] The goal of the safety evaluation for the combination is to determine if there are any increased or unexpected toxicities due to the combination of therapies that would not be expected with either regimen alone (FOLFOX-A or avelumab). As such, a DLT for the combination is any adverse event that is unexpected relative to the known safety profile of the standard and investigational agents in the opinion of the investigator. The phase 2.5 portion of the study will proceed when DLT evaluation period for 6 patients is completed.

Drug Administration

[00673] All subjects: During the course of the study subjects in study Arm A receive intravenous (IV) bevacizumab plus FOLFOX (first line therapy) on Days 1 and 2 of a 2 week cycle as tolerated per investigator discretion for up to a total of 12 cycles in keeping with standard of care treatment guidelines. Subjects in study Arm B receive bevacizumab plus FOLFOX (first line therapy) on Days 2 and 3. After completion of first-line induction therapy, both study arm subjects continue treatment with capecitabine plus bevacizumab on a 2-week cycle in the maintenance period. Body surface area (BSA) and dosing are calculated based on the weight recorded within 2 days prior to avelumab dosing.

[00674] Administration of Bevacizumab plus mFOLFOX6 (Induction Therapy):

[00675] Bevacizumab 5 mg/kg IV over 30 - 90 min on day 1 (infusion rate will be dependent on rate escalation tolerance) (Arm A) or 2 (Arm B),

[00676] Oxaliplatin 85mg/m 2 IV over 2 hours on day 1 (Arm A) or 2 (Arm B),

[00677] Leucovorin* 400mg/m 2 IV over 2 hours on day 1 (Arm A) or 2 (Arm B),

[00678] 5-FU* 400 mg/m 2 IV bolus on day 1 (Arm A) or 2 (Arm B),

[00679] 5-FU* 2400 mg/m 2 r over 46 hours to start on day 1 (Arm A) or 2 (Arm B).

[00680] 5-Fluorouracil and leucovorin should be administered separately to avoid the formation of a precipitate. Per package insert, leucovorin is administered first.

[00681] Administration of Capecitabine plus Bevacizumab (Maintenance Therapy):

[00682] Capecitabine 625 mg/m 2 twice a day every day by mouth. Patients are instructed to take capecitabine with water within 30 minutes after a meal. Doses are rounded to the nearest dose achievable without splitting pills. [00683] Bevacizumab 5 mg/kg IV over 30 - 90 min on day l(Arm A) or 2 (Arm B), repeat every 14 days (infusion rate dependent on rate escalation tolerance)

[00684] Bevacizumab rate escalation

The first dose of bevacizumab is given over 90 minutes. If the dose is well tolerated with no infusion reactions, the rate is increased for the second dose, which is given over 60 minutes. If the second dose is well tolerated, the third dose and all subsequent doses are given over 30 minutes. If there is an initial infusion reaction at 90 or 60 minute infusion times, but subsequent doses are well-tolerated, the rate is escalated at the investigator's discretion.

[00685] Lead-in, Arm A crossovers and Arm B subjects: Subjects in the lead in portion of the study and subjects in Arm B additionally receive the study drugs, Avelumab and Ad-CEA vaccine, during the induction treatment period as well as during the maintenance phase of therapy. Avelumab and Ad-CEA vaccine are given on day 1 and FOLFOX + bevacizumab are given on day 2. Subjects in arm A that progress are offered the opportunity to receive Ad-CEA vaccine and avelumab in combination with a standard chemotherapy regimen

[00686] Avelumab administration: As a routine precaution, subjects enrolled in this trial are observed for 1 hour post infusion, in an area with resuscitation equipment and emergency agents. At all times during avelumab treatment, immediate emergency treatment of an infusion-related reaction or a severe hypersensitivity reaction according to institutional standards must be assured. In order to treat possible anaphylactic reactions, for instance, dexamethasone 10 mg and epinephrine in a 1 : 1000 dilution or equivalents are available along with equipment for assisted ventilation.

[00687] Subjects receive intravenous infusion of avelumab over 1 hour (-10 minutes / +20 minutes, i.e., 50 to 80 minutes) as applicable at a dose of 10 mg/kg.

[00688] Premedication with an antihistamine and with acetaminophen approximately 30 to 60 minutes prior to each dose of avelumab is mandatory (for example, 25-50 mg diphenhydramine and 500-650 mg acetaminophen i.v. or oral equivalent). This regimen is modified based on local treatment standards and guidelines, as appropriate.

[00689] The dose of avelumab is calculated based on the weight of the subject obtained within 2 days prior to avelumab administration. Subjects receive avelumab at the appropriate schedule until confirmed progression, unacceptable toxicity, or any criterion for withdrawal from the therapy.

[00690] Avelumab is given prior to IV chemotherapy during each cycle, when possible.

[00691] Ad-CEA vaccine administration: Ad-CEA vaccine is administered as subcutaneous injection on Day 1 of specific cycles, prior to avelumab. Ad-CEA is administered on cycles 1,2, and 3; 5, 7, and 9; and every 6 cycles thereafter.

[00692] Ad-CEA is provided in a frozen state in a 2 ml vial with a fill volume of 1 ml of extractable vaccine which contains 5xl0 n total virus particles. The product is stored at -20°C until used and immediately thawed before use. [00693] To administer 5x10 n virus particles by subcutaneous injection: 1.0 niL of contents from the previously thawed, supplied Ad-CEA is withdrawn from the vial and administered

subcutaneously to the patient without any further manipulation.

[00694] Lead-in and Arm B Induction and Maintenance: During the induction and maintenance phases (FOLFOX plus bevacizumab, and capecitabine plus bevacizumab, respectively), avelumab are given on day 1 of each cycle (+/- 2 days for logistical reasons).

[00695] Crossovers from Arm A: Subjects that have progressed during the induction phase on Arm A are given the option to cross over to avelumab in combination FOLFOX in order to exploit the synergistic effects of the combination therapy. Subjects on Arm A who have progressed during the maintenance phase are given the option to cross over to avelumab in combination with either capecitabine + bevacizumab or capecitabine+bevacizumab+oxaliplatin based on investigator choice. Subjects that cross over are treated on a 2 week cycle. Avelumab is given on day 1 of each cycle (+/- 2 days for logistical reasons are allowed) and prior to the other agents to avoid giving steroids prior to avelumab.

[00696] Cross-over is not offered in the case of rapid or symptomatic progression due to the need for implementation of a standard option with known efficacy.

[00697] Clinical outcomes in Patients with Colorectal Cancer: Progression Free survival (PFS): 18 patients were enrolled on the single-agent phase I study of avelumab with mCRC at the lOmg/kg dose level. Of these 18, 14 had reached the first restaging for evaluation. Of the 14 evaluable patients, 3 remain on study without evidence of progression for >100 days. Two of these patients have had stable disease for >200 days. Of the remaining evaluable patients (n = 11), 9 have had disease progression within 85 days of initial treatment. The remaining 2 patients came off study for adverse events (transaminase elevation and infusion reaction, respectively). FIG. 54 illustrates the outcomes to date for patients with CRC treated with avelumab

(MSB0010718C) by Response Evaluation Criteria for Solid Tumors (RECIST) criteria.

[00698] CEA response: One patient with mCRC on the phase I study had a remarkable decrease in serum CEA. At baseline his CEA was 267. After starting avelumab, he had a rapid and consistent fall in CEA over the following 5 months with stable disease on scans. He has now been on study for over 11 months with stable disease on scans. Notably, a biopsy of one of his retroperitoneal lymph nodes, being followed as measurable disease on CT scan, revealed "fibroadipose tissue with focal chronic inflammation. No tumor seen." This patient still has elevated CEA in the range of 22-41 that has been holding in this range for 5 months. FIG. 55 demonstrates the change in this patient's CEA.

[00699] Immune data: An analysis of immune cell subset changes from baseline to day 15 and day 43 was performed on PBMC samples from the first 23 patients enrolled on the phase I study of avelumab on clinical trial 13-C-0063 (NCTO 1772004), which included patients at 4 dose levels (1, 3, 10, and 20mg/kg). There was no clear pattern of change (defined as 20% increase or decrease) pre- versus post-treatment in absolute lymphocyte count. A panel of markers was used to define >50 immune cell subsets by flow-cytometry at the same intervals. There were no significant changes in any of the immune cell subsets. Notably, there was no decrease in the number of circulating immune cells expressing PD-L1 (including B lymphocytes and dendritic cells).

[00700] Avelumab Phase I Safety The dose escalation portion of clinical trial 13-C-0063 (NCT01772004) was completed. 4, 14 (1 withdrew before treatment), 12 and 21 patients, respectively, were enrolled into 4 escalating dose levels: 1 mg/kg, 3 mg/kg, 10 mg/kg, and 20 mg/kg. The maximum tolerated dose was not exceeded. At the 20 mg/kg dose level, 1 dose limiting toxicity was experienced out of 6 patients treated. This DLT was associated with a significant anti-tumor response and was attributed to drug because of the possibility that it was an immune-related adverse event (with transient elevated creatine kinase, muscle weakness, myalgia, transaminitis, myocarditis with elevated troponin and inferiolateral ST-elevation on ECG). To date, 80 patients have been enrolled. 55pts (71.2%) have come off-study: 43 (53.7%) for progression and 6 (7.5 %) for toxicity. Grade 3 AEs attributable to drug comprised of 7 laboratory abnormalities (4 increased liver transaminases, 1 decreased lymphocytes, 1 elevated creatine kinase and 1 elevated lipase) in 5 patients and symptomatic myositis requiring intervention in 2 other patients.

[00701] Pharmacokinetics and Receptor Occupancy data: Data from 25 patients was evaluable for PK and RO analysis. Median time to maximum concentration was approximately 1.5 to 2 hours after infusion (for all doses with a linear PK). Half-life was 63.4, 80.7, 93.9 and 115.1 hours for dose levels 1, 2, 3 and 4 respectively, measured by ELISA. Target RO data was available for 13 patients, measured by PD-L1 binding on peripheral leucocytes via flow cytometry. Mean RO was 75.7, 93.8 and 93.2% for dose levels 1, 2 and 3 respectively.

Avelumab: Adverse Drug Reactions (ADRs) Requiring Treatment Discontinuation or Modifications (Note: standard of care therapy will be continued during any AE related delay or discontinuation of avelumab)

[00702] Any Grade 4 ADRs require treatment discontinuation except for single laboratory values out of normal range that are unlikely related to trial treatment as assessed by the investigator, do not have any clinical correlate, and resolve within 7 days with adequate medical management.

[00703] Any Grade 3 ADRs require treatment discontinuation except for any of the following:

[00704] Transient (< 6 hours) Grade 3 flu-like symptoms or fever, which is controlled with medical management.

[00705] Transient (< 24 hours) Grade 3 fatigue, local reactions, headache, nausea, emesis that resolves to < Grade 1. [00706] Single laboratory values out of normal range (excluding > Grade 3 liver function test increase) that are unlikely related to trial treatment according to the investigator, do not have any clinical correlate, and resolve to < Grade 1 within 7 days with adequate medical management.

[00707] Tumor flare phenomenon defined as local pain, irritation, or rash localized at sites of known or suspected tumor.

[00708] Any Grade > 3 drug-related amylase or lipase abnormality that is not associated with symptoms or clinical manifestations of pancreatitis does not require dose delay. The Study Medical

Monitor should be consulted for such Grade > 3 amylase or lipase abnormalities.

[00709] Increases in ECOG performance status > 3, which do not resolve to < 2 by cycle Day 14 of the following cycle (infusions should not be given on the following cycle, if the ECOG performance status is > 3 on the day of study drug administration).

[00710] Any Grade 2 ADR should be managed as follows:

[00711] If a Grade 2 ADR resolves to Grade < 1 by the last day of the current cycle, treatment may continue.

[00712] If a Grade 2 ADR does not resolve to Grade < 1 by the last day of the current cycle, infusions should not be given on the following cycle. If at the end of the following cycle, the event has not resolved to Grade 1, the subject should permanently discontinue treatment with avelumab ADR (except for hormone insufficiencies, that can be managed by replacement therapy; for these hormone insufficiencies, up to 2 subsequent doses may be omitted).

[00713] Upon the second occurrence of the same Grade 2 ADR (except for hormone insufficiencies that can be managed by replacement therapy) in the same subject, treatment with avelumab is permanently discontinued.

[00714] Infusion-related reactions, hypersensitivity reactions (Grades 1 to 4), and tumor lysis syndrome should be handled.

[00715] Recommendations for the management of avelumab infusion related reactions: Infusion of avelumab is stopped in case of > Grade 2 hypersensitivity, inflammatory response, or anaphylactic reaction. The treatment recommendations for infusion-related reactions, severe hypersensitivity reactions, and tumor lysis syndrome according to the NCI are outlined in in the Table 13 below. All infusion-related reactions, occurring during study drug infusion or after completion of the study drug administration, are reported as AESIs or SAEs in case any serious criterion is met. Table 13

[00716] Once the avelumab infusion rate has been decreased by 50% or interrupted due to an infusion related reaction, it must remain decreased for all subsequent infusions. If the subject has a second infusion-related reaction Grade > 2 on the slower infusion rate, the infusion is stopped and the subject is removed from avelumab treatment. If a subject experiences a Grade 3 or 4 infusion- related reaction at any time, the subject must discontinue avelumab. If an infusion reaction occurs, all details about drug preparation and infusion must be recorded.

[00717] Management of Immune Related Adverse Events (irAEs): Since inhibition of PD-L1 stimulates the immune system, irAEs may occur. Treatment of irAEs is mainly dependent upon severity (NCI-CTCAE grade):

[00718] Grade 1 to 2: treat symptomatically or with moderate dose steroids, more frequent monitoring

[00719] Grade 1 to 2 (persistent): manage similar to high grade AE (Grade 3 to 4)

[00720] Grade 3 to 4: treat with high dose corticosteroids

[00721] Treatment of irAEs should follow guidelines set forth in the tables below. Table 14. Management of Gastrointestinal irAEs

Table 15. Management of Dermatological irAEs

Grade of Pneumonitis Management Follow-up

(NCI-CTCAE v4)

Grade 2 Delay avelumab therapy Re-image every 1 to 3 days

Mild to moderate new symptoms Pulmonary and Infectious If improves:

Disease consults When symptoms return to near Monitor symptoms daily, baseline, taper steroids over at least 1 month and then resume consider hospitalization avelumab therapy and consider 1.0 mg/kg/day prophylactic antibiotics

If not improving after 2 weeks or methylprednisolone i.v. or oral

worsening:

equivalent

Consider bronchoscopy, lung Treat as Grade 3 to 4 biopsy

Grade 3 to 4 Discontinue avelumab therapy If improves to baseline:

Severe new symptoms; New / Hospitalize Taper steroids over at least 6 worsening hypoxia; life- Pulmonary and Infectious weeks

If not improving after 48 hours threatening Disease consults

or worsening:

2 to 4 mg/kg/day

methylprednisolone i.v. or i.v. Add additional

immunosuppression (for equivalent example, infliximab,

Add prophylactic antibiotics for cyclophosphamide, i.v.

immunoglobulin, or

opportunistic infections mycophenolate mofetil) Consider bronchoscopy, lung

biopsy

Table 17. Management of Hepatic irAEs

Grade of Liver Test Elevation Management Follow-up

(NCI-CTCAE v4)

Grade 1 Consider delay of avelumab Re-image at least every 3 weeks

Grade 1 AST or ALT > ULN to therapy If worsens:

3.0 x ULN and / or total bilirubin Monitor for symptoms every 2 to Treat as Grade 2 or Grade 3 to 4 > ULN to 1.5 x ULN 3 days

Consider Pulmonary and

Infectious Disease consults Grade of Liver Test Elevation Management Follow-up

(NCI-CTCAE v4)

Grade 2 Delay avelumab therapy If returns to baseline:

AST or ALT > 3.0 to < 5 x ULN Increase frequency of monitoring Resume routine monitoring, and / or total bilirubin > 1.5 to < to every 3 days resume avelumab therapy

If elevations persist > 5 to 7 days 3 x ULN

or worsen:

0.5 to 1 mg/kg/day

methylprednisolone or oral equivalent and when LFT returns to Grade 1 or Baseline, taper steroids over at least 1 month, consider prophylactic antibiotics for opportunistic infections, and resume avelumab therapy

Grade 3 to 4 Discontinue avelumab therapy If returns to Grade 2:

AST or ALT > 5 x ULN and / or Increase frequency of monitoring Taper steroids over at least 1 total bilirubin > 3 x ULN to every 1 to 2 days month

If does not improve in > 3 to 5 1.0 to 2.0 mg/kg/day

days, worsens or rebounds: methylprednisolone i.v. or i.v.

equivalent Add mycophenolate mofetil 1 gram (g) twice daily

Add prophylactic antibiotics for If no response within an opportunistic infections additional 3 to 5 days:

Consult gastroenterologist consider other

Consider obtaining MRI/CT scan immunosuppressants per local guidelines

of liver and liver biopsy if

clinically warranted

Table 18. Management of Endocrine irAEs

Endocrine Disorder Management Follow-up

Symptomatic endocrinopathy Evaluate endocrine function If returns to baseline:

Consider pituitary scan Resume routine monitoring, Symptomatic with abnormal lab / resume avelumab therapy

If elevations persist > 5 to 7 days pituitary scan:

or worsen:

Delay avelumab therapy

0.5 to 1 mg/kg/day

1 to 2 mg/kg/day

methylprednisolone or oral methylprednisolone i.v. or by

equivalent and when LFT returns mouth equivalent

to Grade 1 or Baseline, taper

Initiate appropriate hormone steroids over at least 1 month, therapy consider prophylactic antibiotics

No abnormal lab / pituitary MRI for opportunistic infections, and scan but symptoms persist: resume avelumab therapy

Repeat labs in 1 to 3 weeks /MRI

in 1 month

Grade 3 to 4 Delay or discontinue avelumab therapy per section "Avelumab:

Suspicion of adrenal crisis (for Adverse Drug Reactions (ADRs) Requiring Treatment example, severe dehydration, Discontinuation or Modifications"

hypotension, shock out of

Rule out sepsis

proportion to current illness)

Stress dose of i.v. steroids with mineralocorticoid activity i.v. fluids Consult endocrinologist

If adrenal crisis ruled out, then treat as above for symptomatic endocrinopathy

[00722] Active Immunotherapy with Ad-CEA vaccine will be Discontinued for:

[00723] Life-threatening anaphylactic reactions related to active immunotherapy

[00724] DLT related to active immunotherapy with Ad-CEA as follows: a) If one or more patients develop a Grade 4 allergic reaction without a clear attributable cause, other than study vaccine, b)

Death not attributed to disease

[00725] Disease progression (by RECIST criteria). Patients are offered referral to a medical oncologist for discussion of other treatment options, and for continued medical care.

[00726] Disease progression prior to completing the study immunizations: In the event that a patient undergoes reimaging studies prior to the completion of their 6 study immunizations and is found to have disease progression, they are permitted to continue on the study as long as the progression has been 50% or less by RECIST criteria. Dose delays/Modifications - Bevacizumab

Hypertension

[00727] Bevacizumab is administered unless SBP< 150 and DBP < 90. Management of hypertension is outlined below.

[00728] Mild/Moderate Hypertension: For SBP > 140 and < 210 mm Hg or DBP > 90 and < 120 mm Hg that is sustained over at least a two week period, initiate or adjust anti -hypertensive therapy. Bevacizumab must be delayed until the blood SBP< 150 and DBP < 90.

[00729] Severe Hypertension: For SBP > 210 mm Hg or DBP > 120 mm Hg but without end organ damage, begin anti -hypertensive therapy. Bevacizumab must be delayed until blood SBP< 150 and DBP < 90.

[00730] Hypertensive Urgency or Emergency: Bevacizumab therapy will be permanently discontinued in the presence of hypertensive urgency (DBP >120 with evidence of optic disc edema or progressive end-organ complications) or hypertensive emergency (SBP > 210 and DBP > 120 presenting with headaches, blurred vision, or focal neurological symptoms, or papilledema). Only grade 4 hypertension will require expedited reporting to the NCI-IRB.

Thrombotic Events

[00731] Deep Vein Thrombosis: Bevacizumab will not be administered in the presence of deep vein thrombosis. Anticoagulation may be used and bevacizumab may be restated when thrombosis is resolved.

[00732] Arterial Thromboembolic Events: Bevacizumab will be permanently discontinued in the presence of new arterial thrombotic disease, including but not limited to, myocardial infarct due to coronary thrombosis and ischemic stroke.

[00733] Neutropenia: Bevacizumab will be held if the absolute neutrophil count (ANC) is less than 750 cells/μΕ until ANC is again above 750 cells^L. Filgrastim or pegfilgrastim may be scheduled to individual patient requirement in order to maintain a neutrophil count targeted to meet or exceed 750 cells/nL.

[00734] Hemorrhage

[00735] Life Threatening: In the case of hemoptysis, hematemesis, hematochezia, intracranial hemorrhage or any significant blood loss, bevacizumab will not be administered.

[00736] Proteinuria: If urine dipstick indicates >2+ proteinuria, bevacizumab administration will be delayed pending the results of a 24-hour urine collection for protein. If the 24-hour urine protein is >2 grams, bevacizumab will be delayed for up to 6 weeks, until the proteinuria is <2+ or the 24 -hour urine protein is < 1 grams/24 hr.

[00737] Liver Function Abnormalities: Bevacizumab should be withheld in the event of > Grade 3 LFT elevations and should not resume until the abnormalities have recovered to < Grade 1 (with the exception of hyperbilirubinemia attributable to protease inhibitor therapy, in which case the total bilirubin must recover to < 7.5 mg/dL and the direct fraction < 0.7 mg/dl.) If LFT elevations recur with re-treatment, bevacizumab should be permanently discontinued. Liver function tests included are the transaminases alanine aminotransferase (ALT, SGPT), aspartate aminotransferase (AST, SGOT), alkaline phosphatase, and bilirubin. Elevated bilirubin due to protease inhibitor therapy will not require treatment modification.

[00738] Allergic Reaction: In case of flushing, shortness of breath, facial edema, headache, chills, back pain, tightness of the chest and throat, and/or hypotension, fever or rash, the infusion should be suspended until the patient is assessed until the events have subsided. For grade 3 or 4 allergic reactions, bevacizumab will be permanently discontinued.

[00739] Surgical or periodontal procedures: If there is need for an elective major surgical or periodontal procedure, bevacizumab should be held beginning at least 6 weeks prior to the procedure and must not be resumed before 4 weeks after the surgical procedure. For urgent or emergent surgery or endoscopic procedures, bevacizumab will be held for 4 weeks after the procedure. Longer delays may be necessary, if clinically indicated, in order to insure that adequate healing has taken place prior to bevacizumab resumption. For minor procedures (i.e. interventional radiology diagnostic procedures), bevacizumab will be held only if there is evidence that healing is compromised.

[00740] Reversible Posterior Leukoencephalopathy Syndrome (RPLS): Bevacizumab should not be administered to patients with signs or symptoms of RPLS. Evaluation should include neurologic evaluation, ocular examination, head MRI, and blood pressure assessment. If these clinical criteria are consistent with the diagnosis of RPLS, bevacizumab will be permanently discontinued.

[00741] Gastrointestinal Perforations and Fistula Formation: Bevacizumab will be discontinued in the event of a gastrointestinal perforation or the formation of a fistula.

[00742] Other Grade 3 or 4 Toxicities: Bevacizumab will be held for any other Grade 3 or 4 toxicity thought to be at least possibly related to bevacizumab. Lymphopenia and other toxicities clearly related to HTV or its therapy or to the Kaposi's sarcoma itself will not require dose modification. Grade 3 or higher asymptomatic hyperuricemia, hypophosphatemia, elevated creatinine phosphokinase levels will not result in dose delay.

Dose delays/Modifications - Capecitabine

[00743] Dose adjustments for toxicities per CTCAE as per table. Table 19.

Summary of management of common Capecitabine toxicities

Toxicity Capecitabine dose modification

Grade 4 neutropenia Hold until recovery until < grade 1 then resume one dose level lower*

> Grade 3 febrile neutropenia Hold until resolution of fever and neutropenia to < grade 1. Then resume on dose level lower*

*Dose reductions for capecitabine

1. Starting dose: 625 mg/m 2 BID continuously

2. DL -1 : 470 mg/m 2 BID continuously

3. DL -2: 310 mg/m 2 BID continuously

4. Discontinuation of capecitabine.

Study Calendar

[00744] Weight may be assessed within 2 days prior to avelumab dosing for all cycles

[00745] Cycle 1 day 1 assessments may be performed within 16 days prior to what is indicated [00746] Other day 1 assessments in cycles 2 and beyond may be performed within 1 week prior to indicated time

Table 21

Medications

: 30 day safety visit occurs 4 - 5 weeks after the last administration of study drug regardless of reason for discontinuation. Information may be obtained by telephone if patient refuses visit.

2 May be performed within 16 days prior to initiation of study therapy.

3

Acute Care Panel: Sodium (NA), Potassium (K), Chloride (CL) Total C02 (Bicarbonate), Creatinine, Glucuose, Urea nitrogen, eGFR; Hepatic Panel: Alkaline Phosphatase, ALT/GPT, AST/GOT, Total Bilirubin, Direct Bilirubin; Mineral Panel: Albumin, Calcium, Magnesium (Mg), Phosphorus

4 ACTH: adrenocorticotropic hormone; ANA: anti-nuclear antibody; RF: rheumatoid factor; TSH: Thyroid- stimulating hormone; T4: Free thyroxine.

5 Only urine dipstick required initially. If positive for protein content, a 24 hour urine collection for protein will be performed.

6 Optional biopsy can be performed at any time on study, after first restaging with patient consent. Initial on-study biopsy should be after at least 4 cycles of SOC. A biopsy at the time of disease progression and/or response should be considered as well.

7

Correlative blood samples: blood collected pre-dose : 6 (lOmL) green top sodium heparin tubes for PBMC; 2 (8mL) SST tubes for serum samples.

[00747] Supportive care

[00748] Recommended Management for Severe Hypersensitivity Reaction and Flu-like Symptoms: Management: 1) Epinephrine injection and dexamethasone infusion, 2) Patient should be placed on monitor immediately, 3) Alert intensive care unit (ICU) for possible transfer if required. For prophylaxis of flu-like symptoms, 25 mg indomethacin or comparable NSAID dose (e.g., ibuprofen 600 mg, naproxen sodium 500 mg) may be administered 2 hours before and 8 hours after the start of each dose of avelumab i.v. infusion. Alternative treatments for fever (e.g., acetaminophen) may be given to subjects at the discretion of the investigator.

[00749] Recommendations for the management of tumor lysis syndrome: Since avelumab can induce ADCC, there is a potential risk of tumor lysis syndrome. Should this occur, subjects should be treated as per local guidelines and the management algorithm (FIG. 63).

[00750] General supportive care: Because there is a potential for interaction of capecitabine with other concomitantly administered drugs through the CYP2C9 system, the case report form (CRF) must capture the concurrent use of all other drugs, over-the-counter medications, or alternative therapies. The Principal Investigator should be alerted if the patient is taking any agent known to affect or with the potential to interact with capecitabine. Patients will be transitioned to an acceptable alternative if available. Common examples include aluminum and magnesium hydroxide containing antacids, Coumadin, and phenytoin.

[00751] Diarrhea will be managed with the following regimen: loperamide (4 mg PO) at onset of symptoms, followed by 2 mg loperamide every 2 hours while awake (or 4 mg PO every 4 hours while sleeping) up to a maximum of 16 mg loperamide per day. Additional agents may be used concurrently if loperamide is not adequate to control diarrhea as a single agent.

[00752] Nausea and vomiting will be managed through the use of appropriate simple supportive measures. Recommended premedication for oxalaplatin includes 10-20mg IV dexamethasone and 16mg ondansetron intravenously 30-60 minutes prior to infusion.

[00753] Patients will receive full supportive care per NIH CC guidelines as needed while on this study including blood product support, intravenous hydration, antibiotic treatment and treatment of other newly diagnosed or concurrent medical conditions.

[00754] Patients should be advised to drink plenty of water or take rehydration fluids to avoid dehydration if diarrhea occurs.

Correlative Studies for Research/Pharmacokinetic Studies

[00755] Immunological Studies: Samples from patients enrolled at Georgetown and the Clinical Center will be available for immunologic correlative studies as described below.

[00756] Peripheral blood: Six (lOmL) green top sodium heparin tubes for PBMC; 2 (8mL) SST tubes for serum samples will be drawn on day 1 of Cycle 1, 3, 5, 9 and every 4 cycles thereafter as described in the Table 21. The following assessments are planned in the Laboratory of Tumor Immunology and Biology (LTIB), CCR, NCI for the samples collected.

[00762] Leukocyte subpopulations and immune activation status: Leukocyte subpopulations and immune activation status will be assessed by flow cytometry (FACS) on PBMC. Multiparameter FACS analysis using combination of markers will be performed to characterize leukocyte subpopulations and their functional state such as:

[00763] T-cells subsets and activation state

[00764] T-cells functionality

[00765] PD-1 signaling pathway

[00766] Regulatory T-cells

[00767] Other leukocytes subsets including B-cells, monocytes

[00768] NK cells and related cytotoxicity

[00769] Differentiation stage of T-cells subsets [00770] Myeloid derived suppressor cells and APCs

[00771] Comparison presence of TAA-specific T lymphocytes using TAA-specific peptides will be performed on pre- and post-treatment samples if enough PBMCs are available

[00772] Further exploratory markers related to the mechanism of action of the drug such as soluble

PD-Ll sera level, cytokine profile, and auto-antigen proteomic arrays may be explored.

[00773] T Cell Clonal Expansion Assay: cDNA from PBMC will be amplified using locus specific primers for TCR-β. Previously described methods will be used to map the V region and identify the

J region (Klinger) and identify clonal sequences of interest. Analytics tools will be used to sort and identify clonal populations of interest in the post- versus pre-treatment samples. Correlation of expansion of a clonal TCR population post treatment will be correlated with clinical outcomes.

[00774] Serum soluble factors and ADCC-related side effects (such as in vitro ADCC activity assays, sCD40L and/or sCD27) may be explored.

[00775] Tissue analysis (Optional): Specimens will be collected after 4 cycles of standard of care therapy, and at disease progression and/or response. Remaining tissue from the mandatory samples collected at baseline (archived or fresh) may also be used in this study. Biopsies will be collected using standard techniques which may include which may include CT, cone beam CT, ultrasound, or fusion guided biopsy. Biopsies will be obtained from primary tumor sites and metastatic sites (if applicable). No more than 4 cores may be obtained per site. When possible, the same site of disease should be biopsied on repeat evaluation as was biopsied at baseline.

[00776] Specimens are used for the following studies conducted in the LTIB, CCR, NCI:

[00777] Level of PD-Ll expression will be assessed by immunohistochemistry staining (IHC). Of note, further techniques to evaluate the expression of PD-Ll and/or marker candidates impacting the targeting or contributing to improve its expression may be also investigated if needed.

[00778] Frequency and localization of tumor-infiltrated leukocytes (e.g., CD8, CD4 T-cells, Treg, NK cells, macrophage (Ml/2 profile) by IHC

[00779] Gene expression-based subtyping

[00780] Analysis to be performed in an outside lab, to be determined, using the methods described in Guinney, et al. (The consensus molecular subtypes of colorectal cancer, Nature Medicine, 2015). The clinical and immunologic effects of treatment in the standard and combination arms by subtype will be evaluated. Additional analysis may be performed to evaluate the expression of immune- related genes in tumor tissue pre- and post-treatment in each arm.

Whole Genome / RNA expression / Proteomic Analysis

[00781] FFPE and/or fresh frozen tissue will be sent to NantOmics (Rockville, MD) for analysis of genetic mutations, RNA expression profiling, and proteomic profiling of patient tumor samples from baseline and on treatment. Number of mutations, mutation patterns, RNA expression, and protein expression will be correlated with clinical outcomes in both arms. These analyses will be exploratory and retrospective and used for future hypothesis generation.

[00782] Data collection and Evaluation

[00783] Data will be entered in C3D at all study sites. All data will be kept secure. The PI will be responsible for overseeing entry of data into an in-house password protected electronic system and ensuring data accuracy, consistency and timeliness. The principal investigator, associate

investigators/research nurses and/or a contracted data manager will assist with the data management efforts. All human subjects personally identifiable information (ΡΠ) as defined in accordance to the Health Insurance Portability and Accountability Act, eligibility and consent verification will be recorded. Primary data obtained during the conduct of the protocol will be kept in secure network drives or in approved alternative sites that comply with NIH security standards. Primary and final analyzed data will have identifiers so that research data can be attributed to an individual human subject participant.

[00784] Response Criteria: For the purposes of this study, patients should be re-evaluated for response every 8 weeks. In addition to a baseline scan, confirmatory scans should also be obtained up to 6 (not less than 4) weeks following initial documentation of objective response.

[00785] Response and progression will be evaluated in this study using the new international criteria proposed by the revised Response Evaluation Criteria in Solid Tumors (RECIST) guideline (version 1.1) 73 . Changes in the largest diameter (uni dimensional measurement) of the tumor lesions and the shortest diameter in the case of malignant lymph nodes are used in the RECIST criteria.

[00786] Definitions:

[00787] Evaluable for toxicity: All patients will be evaluable for toxicity from the time of their first treatment with avelumab.

[00788] Evaluable for DLT: The first 6 subjects enrolled enrolled will be evaluable for DLT from the time of their first treatment with avelumab until 28 days have passed. Subjects who do not complete the DLT observation period for reasons other than a DLT will be replaced.

[00789] Evaluable for objective response: Only those patients who have measurable disease present at baseline, have received at least one cycle of therapy, and have had their disease re-evaluated will be considered evaluable for response. These patients will have their response classified according to the definitions stated below.

[00790] Evaluable Non-Target Disease Response: Patients who have lesions present at baseline that are evaluable but do not meet the definitions of measurable disease, have received at least one cycle of therapy, and have had their disease re-evaluated will be considered evaluable for non-target disease. The response assessment is based on the presence, absence, or unequivocal progression of the lesions.

[00791] Disease parameters: [00792] Measurable disease: Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter to be recorded) as >20 mm by chest x-ray, as >10 mm with CT scan, or >10 mm with calipers by clinical exam. All tumor measurements must be recorded in millimeters (or decimal fractions of centimeters).

[00793] Malignant lymph nodes. To be considered pathologically enlarged and measurable, a lymph node must be >15 mm in short axis when assessed by CT scan (CT scan slice thickness recommended to be no greater than 5 mm). At baseline and in follow-up, only the short axis will be measured and followed.

[00794] Non-measurable disease. All other lesions (or sites of disease), including small lesions (longest diameter <10 mm or pathological lymph nodes with >10 to <15 mm short axis), are considered non-measurable disease. Bone lesions, leptomeningeal disease, ascites,

pleural/pericardial effusions, lymphangitis cutis/pulmonitis, inflammatory breast disease, and abdominal masses (not followed by CT or MRI), are considered as non-measurable.

[00795] Note: Cystic lesions that meet the criteria for radiographically defined simple cysts should not be considered as malignant lesions (neither measurable nor non-measurable) since they are, by definition, simple cysts.

[00796] 'Cystic lesions' thought to represent cystic metastases can be considered as measurable lesions, if they meet the definition of measurability described above. However, if non-cystic lesions are present in the same patient, these are preferred for selection as target lesions.

[00797] Target lesions. All measurable lesions up to a maximum of 2 lesions per organ and 5 lesions in total, representative of all involved organs, should be identified as target lesions and recorded and measured at baseline. Target lesions should be selected on the basis of their size (lesions with the longest diameter), be representative of all involved organs, but in addition should be those that lend themselves to reproducible repeated measurements. It may be the case that, on occasion, the largest lesion does not lend itself to reproducible measurement in which circumstance the next largest lesion which can be measured reproducibly should be selected. A sum of the diameters (longest for non-nodal lesions, short axis for nodal lesions) for all target lesions will be calculated and reported as the baseline sum diameters. If lymph nodes are to be included in the sum, then only the short axis is added into the sum. The baseline sum diameters will be used as reference to further characterize any objective tumor regression in the measurable dimension of the disease.

[00798] Non-target lesions. All other lesions (or sites of disease) including any measurable lesions over and above the 5 target lesions should be identified as non-target lesions and should also be recorded at baseline. Measurements of these lesions are not required, but the presence, absence, or in rare cases unequivocal progression of each should be noted throughout follow-up.

[00799] Methods for Evaluation of Measurable Disease: [00800] All measurements should be taken and recorded in metric notation using a ruler or calipers. All baseline evaluations should be performed as closely as possible to the beginning of treatment and never more than 4 weeks before the beginning of the treatment.

[00801] The same method of assessment and the same technique should be used to characterize each identified and reported lesion at baseline and during follow-up. Imaging-based evaluation is preferred to evaluation by clinical examination unless the lesion(s) being followed cannot be imaged but are assessable by clinical exam.

[00802] Clinical lesions: Clinical lesions will only be considered measurable when they are superficial (e.g., skin nodules and palpable lymph nodes) and · 10 mm diameter as assessed using calipers (e.g., skin nodules). In the case of skin lesions, documentation by color photography, including a ruler to estimate the size of the lesion, is recommended.

[00803] Chest x-ray: Lesions on chest x-ray are acceptable as measurable lesions when they are clearly defined and surrounded by aerated lung. However, CT is preferable.

[00804] Conventional CT and MRI: This guideline has defined measurability of lesions on CT scan based on the assumption that CT slice thickness is 5 mm or less. If CT scans have slice thickness greater than 5 mm, the minimum size for a measurable lesion should be twice the slice thickness. MRI is also acceptable in certain situations (e.g. for body scans).

[00805] Use of MRI remains a complex issue. MRI has excellent contrast, spatial, and temporal resolution; however, there are many image acquisition variables involved in MRI, which greatly impact image quality, lesion conspicuity, and measurement. Furthermore, the availability of MRI is variable globally. As with CT, if an MRI is performed, the technical specifications of the scanning sequences used should be optimized for the evaluation of the type and site of disease. Furthermore, as with CT, the modality used at follow-up should be the same as was used at baseline and the lesions should be measured/assessed on the same pulse sequence. It is beyond the scope of the RECIST guidelines to prescribe specific MRI pulse sequence parameters for all scanners, body parts, and diseases. Ideally, the same type of scanner should be used and the image acquisition protocol should be followed as closely as possible to prior scans. Body scans should be performed with breath-hold scanning techniques, if possible.

[00806] PET-CT: At present, the low dose or attenuation correction CT portion of a combined PET-CT is not always of optimal diagnostic CT quality for use with RECIST measurements.

However, if the site can document that the CT performed as part of a PET-CT is of identical diagnostic quality to a diagnostic CT (with Γν and oral contrast), then the CT portion of the PET-CT can be used for RECIST measurements and can be used interchangeably with conventional CT in accurately measuring cancer lesions over time. Note, however, that the PET portion of the CT introduces additional data which may bias an investigator if it is not routinely or serially performed. [00807] Ultrasound: Ultrasound is not useful in assessment of lesion size and should not be used as a method of measurement. Ultrasound examinations cannot be reproduced in their entirety for independent review at a later date and, because they are operator dependent, it cannot be guaranteed that the same technique and measurements will be taken from one assessment to the next. If new lesions are identified by ultrasound in the course of the study, confirmation by CT or MRI is advised. If there is concern about radiation exposure at CT, MRI may be used instead of CT in selected instances.

[00808] Endoscopy, Laparoscopy: The utilization of these techniques for objective tumor evaluation is not advised. However, such techniques may be useful to confirm complete

pathological response when biopsies are obtained or to determine relapse in trials where recurrence following complete response (CR) or surgical resection is an endpoint.

[00809] Tumor markers: Tumor markers alone cannot be used to assess response. If markers are initially above the upper normal limit, they must normalize for a patient to be considered in complete clinical response. Specific guidelines for both CA-125 response (in recurrent ovarian cancer) and PSA response (in recurrent prostate cancer) have been published [JNCI 96:487-488, 2004; J Clin Oncol 17, 3461-3467, 1999; J Clin Oncol 26: 1148-1159, 2008]. In addition, the Gynecologic Cancer Intergroup has developed CA-125 progression criteria which are to be integrated with objective tumor assessment for use in first-line trials in ovarian cancer [JNCI 92: 1534-1535, 2000].

[00810] Cytology, Histology: These techniques can be used to differentiate between partial responses (PR) and complete responses (CR) in rare cases (e.g., residual lesions in tumor types, such as germ cell tumors, where known residual benign tumors can remain).

[00811] The cytological confirmation of the neoplastic origin of any effusion that appears or worsens during treatment when the measurable tumor has met criteria for response or stable disease is mandatory to differentiate between response or stable disease (an effusion may be a side effect of the treatment) and progressive disease.

[00812] FDG-PET: While FDG-PET response assessments need additional study, it is sometimes reasonable to incorporate the use of FDG-PET scanning to complement CT scanning in assessment of progression (particularly possible 'new' disease). New lesions on the basis of FDG-PET imaging can be identified according to the following algorithm: a) Negative FDG-PET at baseline, with a positive FDG-PET at follow-up is a sign of PD based on a new lesion, b) No FDG-PET at baseline and a positive FDG-PET at follow-up: If the positive FDG-PET at follow-up corresponds to a new site of disease confirmed by CT, this is PD. If the positive FDG-PET at follow-up is not confirmed as a new site of disease on CT, additional follow-up CT scans are needed to determine if there is truly progression occurring at that site (if so, the date of PD will be the date of the initial abnormal FDG-PET scan). If the positive FDG-PET at follow-up corresponds to a pre-existing site of disease on CT that is not progressing on the basis of the anatomic images, this is not PD, c) FDG-PET may be used to upgrade a response to a CR in a manner similar to a biopsy in cases where a residual radiographic abnormality is thought to represent fibrosis or scarring. The use of FDG-PET in this circumstance should be prospectively described in the protocol and supported by disease-specific medical literature for the indication. However, it must be acknowledged that both approaches may lead to false positive CR due to limitations of FDG-PET and biopsy resolution/sensitivity.

[00813] Note: A 'positive' FDG-PET scan lesion means one which is FDG avid with an uptake greater than twice that of the surrounding tissue on the attenuation corrected image.

Response criteria:

[00814] Evaluation of Target Lesions:

[00815] Complete Response (CR): Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm.

[00816] Partial Response (PR): At least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum of diameters.

[00817] Progressive Disease (PD): At least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progressions).

[00818] Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum of diameters while on study.

[00819] Evaluation of Non-Target Lesions:

[00820] Complete Response (CR): Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size (<10 mm short axis).

[00821] Note: If tumor markers are initially above the upper normal limit, they must normalize for a patient to be considered in complete clinical response.

[00822] Non-CR/Non-PD: Persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits.

[00823] Progressive Disease (PD): Appearance of one or more new lesions and/or unequivocal progression of existing non-target lesions. Unequivocal progression should not normally trump target lesion status. It must be representative of overall disease status change, not a single lesion increase.

[00824] Although a clear progression of "non-target" lesions only is exceptional, the opinion of the treating physician should prevail in such circumstances, and the progression status should be confirmed at a later time by the review panel (or Principal Investigator).

[00825] Evaluation of Best Overall Response: [00826] The best overall response is the best response recorded from the start of the treatment until disease progression/recurrence (taking as reference for progressive disease the smallest

measurements recorded since the treatment started). The patient's best response assignment will depend on the achievement of both measurement and confirmation criteria.

Table 22. For Patients with Measurable Disease (i.e., Target Disease)

Patients with a global deterioration of health status requiring discontinuation of treatment without objective evidence of disease progression at that time should be reported as "symptomatic deterioration. " Every

effort should be made to document the objective progression even after discontinuation of treatment.

Table 23. For Patients with Non-Measurable Disease (i.e., Non-Target Disease)

Duration of response

[00827] Duration of overall response: The duration of overall response is measured from the time measurement criteria are met for CR or PR (whichever is first recorded) until the first date that recurrent or progressive disease is objectively documented (taking as reference for progressive disease the smallest measurements recorded since the treatment started).

[00828] The duration of overall CR is measured from the time measurement criteria are first met for CR until the first date that progressive disease is objectively documented.

[00829] Duration of stable disease: Stable disease is measured from the start of the treatment until the criteria for progression are met, taking as reference the smallest measurements recorded since the treatment started, including the baseline measurements.

[00830] Progression -Free Survival (PFS) or Time to Progression (TTP): PFS or TTP is defined as the duration of time from start of treatment to time of progression or death, whichever occurs first. Response Review

[00831] An independent review of CT scans and MRIs used in determining response will be undertaken at the end of the study by a collaborator in the Radiology and Imaging Sciences. Images will be reviewed using RECIST 1.1. Initial evaluation of progression should be confirmed with a follow up scan in 4-6 weeks, if clinically reasonable. Date of progression should be the initial date of progression when confirmation is obtained. If no confirmation scan is performed, the initial scan demonstrating progression will determine progression.

[00832] Statistical consideration

[00833] The primary objective of the trial is to determine if there is a difference in time to progression among patients with metastatic colorectal cancer who are treated with standard of care vs. standard of care + anti-PD-Ll monoclonal antibody.

[00834] Based upon results in the literature, patients who would be eligible to be randomized on this trial would be expected to have an estimated 10 month median time to progression with standard of care alone. The goal of this study will be to determine if the use of the anti-PD-Ll monoclonal antibody along with standard of care will result in patients having an increased median time to progression of 18 months. Kaplan-Meier curves and a two-tailed log-rank test will be the primary analysis methods. Assuming exponential time to progression curves, the hazard rate for standard of care alone is 0.0693 or approximately a 7% probability of progressing each month when the median time to progression is 10 months. If it is contemplated that the combination arm will be associated with a median time to progression of 18 months, this corresponds to a hazard rate of 0.0385 and the resulting hazard ratio for the comparison of the two overall actuarial curves would be 1.80.

Following the principals of a phase 2.5 design, to compare these curves and detect a difference with a 0.10 one-tailed log-rank test, a total of 35 evaluable subjects per arm (70 total) will need to be randomized over a one year period and a maximum follow-up of three years, with occurrence of 52 total progressions, in order to have 80% power to compare the curves.

[00835] It is expected that all 76 patients (6 for safety lead-in plus 70 for phase 2.5) can be accrued onto this trial in one year. In order to allow for a very small number of inevaluable patients, the accrual ceiling will be set at 81 patients.

Pharmaceutical Information - AD-CEA Vaccine

[00836] Source -Ad-CEA will be supplied by the manufacturer, Etubics Corporation, through a Cooperative Research and Development Agreement (CRADA). The manufacturing department of Etubics will supply the vaccine, which will be distributed to the sites by Etubics Corporation.

Toxicity

[00837] A Phase I/II clinical trial of ETBX-011 (Ad5 [E1-, E2b-]-CEA(6D)) (IND#14325) that expresses the tumor-associated antigen carcinoembryonic antigen (CEA) has been performed. A summary of the study results on the clinical trial is presented below.

[00838] Schedule, Dosing, and Safety: The primary objective of the Phase Ι/Π dosing trial was to assess safety and a secondary objective was to evaluate CEA-specific immune responses to CEA and to obtain preliminary data on response rate. The study was performed under an FDA-approved IND (IND14325) and registered at ClinicalTrials.gov (NCT01147965). Participants were recruited from oncology clinics at Duke University Medical Center and Medical Oncology Associates, Spokane, WA and provided informed consent that was approved by IRB's. Patients with a histological confirmed diagnosis of metastatic malignancy who were previously treated with standard therapy known to have a possible survival benefit were enrolled into the study. For this study, the carcinoma must have had over expression of CEA as defined by immunohistochemical staining (at least 50% of the tumor with at least moderate intensity of staining) or a carcinoma known to be universally CEA positive i.e. colorectal adenocarcinoma). Patients were not treated until 4 or more weeks after any prior chemotherapy or radiation therapy. They could not have a history of autoimmune disease, serious intercurrent chronic or acute illness, active hepatitis, serologic evidence for HIV infection, or be receiving steroid or immunosuppressive therapy. All patients were >21 years old and had a Karnofsky Performance Score of 70% or higher and a life expectancy of at least 3 months. Pregnant women and nursing mothers were excluded. Dose Limiting Toxicities (DLTs) were defined as any Grade 3 or 4 immediate hypersensitivity reactions, Grade 3 or 4 fever that may possibly be associated with the immunization, Grade >2 autoimmune events except for vitiligo or fever for less than 2 days and less than 101.5 °F, Grade >2 allergic reactions (Grade 2 is defined as generalized urticaria as defined by NCI Common Terminology Criteria for Adverse Events (CTCAE version 4.0), or Grade >3 non-hematologic toxicity. The Ad5 [E1-, E2b-]-CEA(6D) injections were given subcutaneously in the same thigh. The doses were administered every 3 weeks for 3 treatments as follows: Cohort 1 (3 patients) 109 VP in 0.5 ml; Cohort 2 (3 patients): 1010 VP; Cohort 3 (6 patients) 1011 VP. Following establishment of safety in Phase I, 12 additional patients were entered into a Phase Π using the 1011 VP/dose. To evaluate a higher dose, 6 additional patients (cohort 5) received 5x1011 VP/dose.

[00839] Patient Demographics: One patient with CEA expressing bladder and one patient with lung carcinoma was enrolled. Thirty two patients, median age 57.5 (range 38-77) with metastatic colorectal cancer, who had failed a median of three prior chemotherapeutic regimens (range: 2-5), had a median performance status of 90% (range 70-100%), and had a median of three sites of metastatic disease (range 1-5), were enrolled. The majority was able to receive all three

immunizations. Five patients who stopped immunizations early did so due to significant disease progression. The colorectal adenocarcinoma patient demographics compares favorably with previously published studies of patients with chemotherapy-refractory colorectal cancer.

[00840] The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent application, foreign patents, foreign patent application and non-patent publications referred to in this

specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. [00841] Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, application and publications to provide yet further embodiments.

[00842] These and other changes can be made to the embodiments in light of the above -detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.