Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TUMOR-SPECIFIC ADENOVIRUS VECTORS AND THERAPEUTIC USES
Document Type and Number:
WIPO Patent Application WO/2017/075395
Kind Code:
A1
Abstract:
A conditionally replicating adenovirus were generated that can specifically replicate and express therapeutic genes in neuroendocrine tumors. The promoter-specific expression of the adenoviruses is regulated upstream by an INSM1 (insulinoma-associated-1) promoter that is silent in normal adult tissues but active in developing neuroendocrine cells and neuroendocrine tumors. By placing the I NSM 1 -promoter with an insulator and two copies of neuronal restrictive silencer elements in an adenoviral vector, the construct can retain tumor specificity and drive expression of a mutated adenovirus E1A gene (Δ24Ε1Α) and the herpes simplex virus thymidine kinase gene. The I NSM1 -promoter-driven viruses could replicate specifically in the I NSM1 -positive cells and I NSM1 -specific HSV-tk expression in combination with ganciclovir treatment displayed dose-dependent tumor cell-specific killing in insulinomas. When the I NSM1 -promoter driven HSV-tk was combined with Δ24Ε1Α and I NSM 1 p- HSV-tk viruses, the co-infected insulinoma expressed higher levels of HSV-tk and more efficient tumor suppression as compared to the I NSM1 p- HSV-tk virus alone.

Inventors:
LAN MICHAEL (US)
BRESLIN MARY (US)
Application Number:
PCT/US2016/059382
Publication Date:
May 04, 2017
Filing Date:
October 28, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV LOUISIANA STATE (US)
International Classes:
A61K31/7088; A61P35/00
Foreign References:
US20120316225A12012-12-13
US20040191761A12004-09-30
US20070134761A12007-06-14
US20090275042A12009-11-05
US20090238795A12009-09-24
US20080267874A12008-10-30
Other References:
DATABASE Genbank 20 August 1994 (1994-08-20), "Human insulinoma-associated (IA-1) gene , partial cds", XP055380092, Database accession no. U07172
DATABASE Genbank 19 October 2004 (2004-10-19), "Gallus gallus HS4 insulator, partial sequence", XP055380093, Database accession no. U78775
DATABASE Genbank 12 April 2002 (2002-04-12), "Cloning vector pHS4, luciferase reporter vector containing HS4 insulator sequence", XP055380095, Database accession no. AJ277959
Attorney, Agent or Firm:
HAYZER, David J. et al. (US)
Download PDF:
Claims:
CLAIMS

We claim:

1. A neuroendocrine tumor-specific viral expression vector whose genome comprises:

(a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8;

(b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements;

(c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter;

(d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted

bioluminescent reporter molecule; and

(e) at least one nucleotide sequence encoding a 3'-untranslated region,

wherein:

(i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences;

(ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and

(iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

2. The viral expression vector of claim 1 , wherein the nucleotide sequence operably linked to the IRES element encodes the conditionally lethal toxin herpes simplex virus thymidine kinase (HSV-tfr).

3. The viral expression vector of claim 1 , wherein the secreted bioluminescent reporter molecule is a luciferase, a Renilla luciferase, a Metridia luciferase, or a Gaussia luciferase.

4. The viral expression vector of claim 1 , wherein the viral expression vector is a non- replicating Ad5 adenoviral vector. 5. The viral expression vector of claim 1 , wherein the human INSM1 promoter comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region is a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter has at least 95% similarity to SEQ ID NO: 7 and encodes the tumor-selective modified E1A polypeptide Δ24Ε1Α, the IRES element has the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'-untranslated region has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

6. The viral expression vector of claim 5, wherein the nucleotide sequence operably linked to the IRES element has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encodes the herpes simplex virus thymidine kinase ( SV-tk).

7. The viral expression vector of claim 2, wherein the viral expression genome comprises the nucleotide sequence having at least 95% similarity to SEQ ID NO: 11. 8. A composition comprising a first neuroendocrine tumor-specific viral expression vector whose genome comprises:

(a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements;

(c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter;

(d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and

(e) at least one nucleotide sequence encoding a 3'-untranslated region

wherein:

(i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences;

(ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and

(iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells. 9. The composition of claim 8, wherein the nucleotide sequence operably linked to the IRES element encodes the conditionally lethal toxin herpes simplex virus thymidine kinase (HSV- ik).

10. The composition of claim 8, wherein the secreted bioluminescent reporter molecule is a luciferase 2, a Renilla luciferase, a Metridia luciferase, or a Gaussia luciferase.

11. The composition of claim 8, wherein the viral expression vector is a non-replicating Ad5 adenoviral vector. 12. The composition of claim 8, wherein the human INSM1 promoter comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region is a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter has at least 95% similarity to SEQ ID NO: 7 and encodes the tumor-selective modified E1A polypeptide Δ24Ε1Α, an IRES element has the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'- untranslated region has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

13. The composition of claim 12, further comprising the herpes simplex virus thymidine kinase ( SV-tk) having the nucleotide sequence having at least 95% similarity to SEQ ID NO: 9.

14. The composition of claim 2, wherein the viral expression genome comprises the nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 1.

15. The composition of claim 1 , wherein said composition further comprises a

pharmaceutically acceptable carrier.

16. The composition of claim 1 , wherein said composition further comprises a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising:

(a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule;

(b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements;

(c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and

(d) a nucleotide sequence encoding a 3'-untranslated region,

wherein:

(i) said second viral expression vector is competent to infect at least some mammalian cells;

(ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences;

(iii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and

(iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells;

wherein the level of expression of the polypeptide toxin or the polypeptide reporter molecule from the combination of the first and second vectors in a recipient cell is synergistically greater compared to expression from the first or second vector alone.

17. The composition of claim 16, wherein the first and the second viral expression vectors each express the conditionally lethal toxin herpes simplex virus thymidine kinase (HSV-TK).

18. The composition of claim 16, wherein the first and the second viral expression vectors each express a reporter molecule.

19. The composition of claim 16, wherein the first and the second viral expression vectors are each genetically modified non-replicating Ad5 adenoviral vectors.

20. The composition of claim 16, wherein the composition is formulated for directed delivery to a tumor, intravenous delivery to a tumor, or respiratory delivery to a lung tumor. 21. A composition according to any of claims 8-20 comprising a first neuroendocrine tumor- specific viral expression vector and at least one of a second neuroendocrine tumor-specific viral expression vector and a pharmaceutically acceptable carrier.

22. A method of increasing the tumor-specific expression of a polypeptide from a viral vector, said method comprising delivering to a neuroendocrine tumor cell or tumor a first neuroendocrine tumor-specific viral expression vector whose genome comprises:

(a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements;

(c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter;

(d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted

bioluminescent reporter molecule; and

(e) at least one nucleotide sequence encoding a 3'-untranslated region

wherein:

(i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences;

(ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and

(iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

23. The method of claim 22, further comprising delivering to the neuroendocrine tumor cell or tumor a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising:

(a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule;

(b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements;

(c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and

(d) a nucleotide sequence encoding a 3'-untranslated region,

wherein:

(i) said second viral expression vector is competent to infect at least some

mammalian cells;

(ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences;

(iii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and

(iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells;

wherein the level of expression of the at least one of the polypeptide toxin and the polypeptide reporter molecule from the combination of the first and second vectors in the recipient cell or tumor is synergistically greater compared to expression from the first or second vector alone.

24. The method of claim 22, wherein the first and second viral vectors are delivered to the recipient cell or tumor sequentially.

25. The method of claim 22, wherein the first and second viral vectors are co-delivered in a single pharmaceutically acceptable composition.

26. The method of claim 22, wherein in the first neuroendocrine tumor-specific viral expression vector the human INSM 1 promoter comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region is a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter has at least 95% similarity to SEQ ID NO: 7 and encodes the tumor-selective modified E1A polypeptide Δ24Ε1Α, an IRES element has the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'-untranslated region has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10. 27. The viral expression vector of claim 23, wherein the nucleotide sequence operably linked to the IRES element has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encodes the herpes simplex virus thymidine kinase ( SV-tk).

28. The method of claim 27, wherein the first neuroendocrine tumor-specific viral expression genome comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 11.

29. The method of claim 28, wherein in the second neuroendocrine tumor-specific viral expression vector the human INSM 1 promoter comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region is a chicken HS4 β-globin insulator element comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each core element comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encodes the herpes simplex virus thymidine kinase ( SV-tk), and the 3'- untranslated region has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

30. The method of claim 22, wherein at least one of the viral expression vectors thereof encodes a reporter molecule, and the method further comprises observing subsequent expression of the encoded reporter molecule in the patient's tissues as a measure of the presence or the extent of a neuroendocrine tumor.

31. The method of claim 23, wherein the reporter molecule is Gaussia luciferase and a nucleotide sequence encoding said luciferase has at least 95% similarity to SEQ ID NO: 5.

32. The method of claim 24, wherein the first and second neuroendocrine tumor-specific viral expression vectors each comprise a nucleotide sequence encoding a polypeptide toxin and wherein, if said polypeptide toxin is only conditionally lethal, then said method additionally comprises the step of providing conditions that produce the lethal phenotype; whereby cells of the neuroendocrine tumor are selectively killed.

33. The method of claim 34, said method further comprising contacting the neuroendocrine cell with ganciclovir thereby selectively killing cells of the neuroendocrine tumor.

34. A method according to any of claims 22-33, or any combination thereof.

Description:
TUMOR-SPECIFIC ADENOVIRUS VECTORS AND THERAPEUTIC USES

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to and the benefit of U.S. Provisional Application 62/247,229 titled "TUMOR-SPECIFIC ADENOVIRUS VECTORS AND THERAPEUTIC USES" filed October 28, 2015, the entire disclosure of which is incorporated herein by reference.

SEQUENCE LISTING

This application contains a sequence listing filed in electronic form as an ASCII.txt file entitled "2222272100_ST25" created on October 18, 2016. The content of the sequence listing is incorporated herein in its entirety.

TECHNICAL FIELD

The present disclosure is generally related to compositions and methods for enhancing tumor-specific gene expression from a viral vector. The present disclosure is also generally related to methods of inhibiting the proliferation of tumor cells, and especially of insulinoma cells, or their imaging.

BACKGROUND

Insulinoma-associated 1 (INSM1) is a transcriptional repressor protein that is required for the development of the endocrine pancreas, adrenal gland, basal neuronal progenitor cells in the neocortex, and the monoaminergic neurons in the hindbrain (Lan & Breslin (2009) FASEB J. 23: 2024-2033). INSM1 expression is restricted to early fetal development in neuronal and endocrine tissues (Breslin et al., (2003) J. Biol. Chem. 278: 38991-38997; Duggan et al., (2008) J. Comp Neurol. 507: 1497-1520; Goto et al., (1992) J. Biol. Chem. 267: 15252-15257; Mellitzer et al., (2006) EMBO J. 25: 1344-1352; X\e et al., (2002) Genomics 80: 54-61). One striking feature of the INSM1 mRNA is despite its absence in normal adult tissues, it is strongly expressed in tumors of neuroendocrine origin such as small cell lung carcinoma (SCLC), medullablastoma, neuroblastoma, medullary thyroid carcinoma, insulinoma, retinoblastoma, pheochromocytoma, and pituitary tumors (Goto et ai, (1992) J. Biol. Chem. 267: 15252-15257; Lan et ai, (1993) Cancer Res. 53: 4169-4171 ; Taniwaki et ai., (2006) Int. J. Oncol. 29: 567-575). Using a transgenic animal model and in vitro reporter gene assays, the spatial and temporal expression of INSM1 has been demonstrated to be regulated by the 5' 1.7 kilobase pair promoter region (Breslin et al., (2003) J. Biol. Chem. 278: 38991-38997; Li et ai., (1997) Biochem. Biophys. Res.

Commun. 236: 776-781). The 1.7 kbp promoter region has been linked to a suicide gene for delivery into tumor cells. The ability of the INSM1 promoter to drive expression of the herpes simplex virus thymidine kinase gene selectively has been tested in small cell lung cancer (SCLC) cells and in pediatric brain tumors (Pedersen et al., (2006) Cancer Gene Ther. 13: 375-384; Wang et ai, (2009) Hum. Gene Ther. 20: 1308-1318; see also, U.S. Patent Application Publication No. 2005/0037445). Adenoviral vectors are one of the most widely exploited viral delivery systems for gene therapy due to their ability to infect a wide range of host cells and the minimal risk associated with the use of a non-replicating form of the virus. The adenovirus genome is easily manipulated and with the deletion of the E1 and E3 genes allows for the incorporation of up to 7.5 kilobase pairs of exogenous sequence. However, one major drawback of adenovirus is host mediated immunity to the virus. In addition, due to the high liver transduction efficiency following intravenous delivery of adenovirus, the liver is most susceptible to toxic side effects.

SUMMARY

The compositions and methods of the disclosure are suitable for the treatment and/or diagnosis of human neuroendocrine tumors using a combination of a first expression construct comprising the nucleic acid sequence from the human insulinoma-associated 1 (INSM1) promoter, one or more elements selected from neuron restrictive silencer elements and insulator elements, and a suicide or toxin-encoding element for treatment of tumors and/or a reporter gene for visualization or detection. A second expression construct comprises a modified adenovirus E1A region that in combination with the first construct results in enhanced tumor-specific expression of the toxin polypeptide or the reporter gene. The construct may be linked directly with a reporter gene for diagnosis of neuroendocrine tumors.

One aspect of the disclosure, therefore, encompasses embodiments of a

neuroendocrine tumor-specific viral expression vector whose genome comprises: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

Another aspect of the disclosure encompasses embodiments of a composition comprising a first neuroendocrine tumor-specific viral expression vector whose genome comprises: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

In some embodiments of this aspect of the disclosure, the composition can further comprise a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and (d) a nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said second viral expression vector is competent to infect at least some mammalian cells; (ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences; (iii) said INSM 1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells; wherein the level of expression of the polypeptide toxin or the polypeptide reporter molecule from the combination of the first and second vectors in a recipient cell is synergistically greater compared to expression from the first or second vector alone.

Yet another aspect of the disclosure encompasses embodiments of a method of increasing the tumor-specific expression of a polypeptide from a viral vector, said method comprising delivering to a neuroendocrine tumor cell or tumor a first neuroendocrine tumor- specific viral expression vector whose genome comprises: (a) a human Insulinoma- associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'- untranslated region wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

In some embodiments of this aspect of the disclosure, the method can further comprise delivering to the neuroendocrine tumor cell or tumor a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and (d) a nucleotide sequence encoding a 3'- untranslated region, wherein: (i) said second viral expression vector is competent to infect at least some mammalian cells; (ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences; (iii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells; wherein the level of expression of the at least one of the polypeptide toxin and the polypeptide reporter molecule from the combination of the first and second vectors in the recipient cell or tumor is synergistically greater compared to expression from the first or second vector alone.

In some embodiments of this aspect of the disclosure, the first and second neuroendocrine tumor-specific viral expression vectors can each comprise a nucleotide sequence encoding a polypeptide toxin and wherein, if said polypeptide toxin is only conditionally lethal, then said method additionally can comprise the step of providing conditions that produce the lethal phenotype; whereby cells of the neuroendocrine tumor are selectively killed.

BRIEF DESCRIPTION OF THE DRAWINGS

Further aspects of the present disclosure will be more readily appreciated upon review of the detailed description of its various embodiments, described below, when taken in conjunction with the accompanying drawings.

Fig. 1 schematically illustrates the oncolytic and non-oncolytic adenoviral vectors of the disclosure, and their constituent regions.

Fig. 2 illustrates the nucleotide sequence of an INSM1 promoter (-1661-+40 bp) having the sequence SEQ ID NO: 1.

Fig. 3 illustrates the nucleotide sequence of a pair of tandem nicotinic acetylcholine receptor (nAchR) neuronal restrictive silencer elements (NRSEs) having a 5' terminal Bglll restriction site and a 3' Hindi 11 restriction site and having the nucleotide sequence SEQ ID NO: 2.

Fig. 4 illustrates the nucleotide sequence of an HS4 insulator sequence

(approximately 1.2kb) having the nucleotide sequence SEQ ID NO: 3.

Fig. 5 illustrates the nucleotide sequence of an HS4 core sequence (approximately

250 bp) having the nucleotide sequence SEQ ID NO: 4. Fig. 6 illustrates the nucleotide sequence encoding a Gaussia bioluminescent reporter having a 5' terminal Hindlll restriction site and a 3' NotI restriction site and having the nucleotide sequence SEQ ID NO: 5.

Fig. 7 illustrates the nucleotide sequence of an IRES element having the nucleotide sequence SEQ ID NO: 6.

Fig. 8: illustrates the nucleotide sequence encoding a Δ24Ε1Α polypeptide having the nucleotide sequence SEQ ID NO: 7.

Fig. 9 illustrates the amino acid sequence (SEQ ID NO: 8) of a human E1A polypeptide.

Fig. 10 illustrates the nucleotide sequence encoding a thymidine kinase (TK) having the nucleotide sequence SEQ ID NO: 9.

Fig. 1 1 illustrates the nucleotide sequence of a 3'-untranslated region having the nucleotide sequence SEQ ID NO: 10.

Figs. 12A-12E illustrate viral replication and protein expression.

Fig. 12A is a graph illustrating viral replication in I NSM1 -positive rat insulinoma cells

(RIN). RIN cells were seeded in a 6-well plate at a density of 2*10 6 cells per well and infected with a viral concentration of 10 MOI or 200 MOI. After incubation for 3 days, cells and medium were collected and the viral particles were released into the medium by freeze- thaw lysis. The isolated viruses were then titered by immunostaining (n=6). To examine the expression of viral proteins (HSV-tk and E1A), Ad-INSM1 p- HSV-tk and Ad-INSM1 p-E1A- HSV-tk were used to infect RIN, MIN, β-TCI (I NSM1 -positive insulinomas), and Panc-1 (I NSM1 -negative pancreatic epithelioid carcinoma) cells at 50 MOI. After incubation for 48 hours, total protein extracts were isolated from each cell line for HSV-tk and E1A analyses.

Fig. 12B is a digital image of a gel analysis for Ad-INSM1 p-E1A-HSV-tk.

Fig. 12C is a digital image of a gel analysis for Ad-INSM1 p-HSV-tk (K5).

Fig. 12D is a digital image of a gel analysis for Combination viruses.

Fig. 12E is a digital image of a gel analysis for CAR expression in uninfected cells. Figs. 13A-13D illustrate that the oncolytic virus in combination with Ad-INSM1 p-HSV- tk increased efficacy of tumor cell killing. An MTS colorimetric assay was conducted to measure the cell survival with 3 insulinoma cell lines: RIN, MIN, β-TCI (INSM1 +

insulinomas), and Panc-1 (INSM1 " pancreatic epithelioid carcinoma).

Figs. 13A is a graph illustrating cells treated with Ad-INSM1 p-Luc2.

Figs. 13B is a graph illustrating cells treated with Ad-INSM1 p-E1A-HSV-tk

Figs. 13C is a graph illustrating cells treated with Ad-IMSM 1 p-HSV-tk (K5)

Figs. 13D is a graph illustrating cells treated with a combination of Ad-IMSM1 p-HSV- tk and Ad-INSM1 p-E1A-HSV-tk viruses. Cells were first treated with virus ranging from 0- 100 MOI. Three days after infection, cells were treated with 100 μΜ GCV for five additional days. Cell survival was measured by MTS colorimetric analysis at 490 nm. (n=6).

Figs. 14A-14D illustrate that the Ad-INSM1 p-HSV-tk virus in combination with the oncolytic virus kills tumor cells in a GCV dose-dependent manner. An MTS assay was conducted with three insulinoma cell lines: RIN, MIN, β-TCI (all INSM1 + insulinoma), and Panc-1 (INSM1 " pancreatic epithelioid carcinoma).

Figs 14A is a graph illustrating cells treated with Ad-INSM1 p-Luc2.

Figs 14B is a graph illustrating cells treated with Ad-INSM1 p-E1A-HSV-tk.

Figs 14C is a graph illustrating cells treated with Ad-IMSM 1 p-HSV-tk (K5).

Figs 14D is a graph illustrating cells treated with a combination of Ad-IMSM1 p-HSV- tk and Ad-INSM1 p-E1A-HSV-tk viruses. Cancer cells were infected with 50 MOI viruses for 3 days, after which 0-200 μΜ ganciclovir was added for five additional days. Cell survival was measured by MTS colorimetric analysis at 490 nm. (n=6).

Figs. 15A-15C illustrate that a combination virus treatment in an in vivo

subcutaneous mouse xenograft tumor model.

Figs. 15A is a graph illustrating blood glucose levels in RIN cells pre-treated with 5 MOI Ad-INSM1-Luc2 for 24 hours and then injected into the right flank of 6-8 week old Nu/Nu mice. After 48 hours, Ad-INSM 1-Luc2 (10 9 ifu) or a combination of Ad-INSM1-HSV-tk and Ad-INSM1-E1A-HSV-tk (5* 10 8 ifu each) were injected directly into the tumor site (marked after tumor cell injection). GCV treatment was started 3 days after injection (arrow) to allow for viral replication to occur inside the tumor cells. Blood glucose was measured daily via tail vein.

Fig. 15B is a digital image illustrating luminescent activity of the tumor in a representative mouse treated with either Ad-INSM1-Luc2 measured at days 3, 6, 9, and 12.

Fig. 15C is a digital image illustrating luminescent activity of the tumor in a representative mouse treated with a combination of Ad-IMSM1-HSV-tk and Ad-INSM1-E1A- HSV-tk was measured at days 3, 6, 9, and 12.

Fig. 16 is a graph illustrating pre-infection with the combination virus in an in vivo subcutaneous mouse xenograft tumor model. RIN cells were pre-treated with 10 MOI of Ad- INSM1 p-Luc2 or combination of Ad-INSM1 p-HSV-tk and Ad-INSM1 p-E1A-HSV-tk viruses (5 MOI each). After 24 hours of incubation, 10 9 cells were injected subcutaneously into the right flank of 4 week old Nu/Nu mice. GCV treatment was started 3 days after injection (arrow) to allow for viral replication to occur inside the tumor cells. Blood glucose was measured daily via tail vein. (n=5).

Fig. 17 is a series of digital images illustrating the specificity, un-modified INSM1- promoter driven Ad-INSM1 p-/.i/c2 (1 *10 9 ) was injected intravenously into non-tumor bearing Nu/Nu mice and the luciferase signal was measured from days 2 to 28 post injection: Panel A: The first generation; Panel B: second generation modified I NSM1 -promoter driven Ad- INSM1 p- -L c2 (1 * 10 9 ) do not show luciferase activity as INSM1 promoter retains its specificity; Panel C: For the NE tumor imaging, the modified Ad-I NSM 1 p-/.i c2 virus was pre- infected with H1 155 NE lung tumor cells (50 MOI) and injected subcutaneously into the right hind flank of nude mice (n=3). After one week, the tumor growth was evidenced and imaged to show the modified INSM1 promoter specificity. Mouse tumor was faced down for luciferase imaging.

Figs. 18A-18C illustrate Ad-I NSM 1 p-/Wef vector expressed Metridia luciferase specifically in INSM1-positive cell lines. An increasing Ad-INSM1 p-/Wef concentration (0-50 MOI) and a constant Ad-SV40-/_i c2 concentration (5 MOI) was used to infect INSM 1- positive (solid lines) and -negative (dot lines) cell lines in culture.

Fig. 18A is a graph illustrating Ad-INSM1 p-/Wef vector expressed Metridia luciferase expression specifically in NE lung cancer H1 155, UMC-1 1 , H82, and lung adenosquamous carcinoma H596 cells.

Fig. 18B is a graph illustrating Ad-INSM1 p-/Wef vector expressed Metridia luciferase expression specifically in Neuroblastoma SK-N-Be(2) and SH-SY5Y, retinoblastoma WERI- Rb-1 , pheochromocytoma PC-12, medulloblastoma D283 Med, and cervical

adenocarcinoma HeLa cells.

Fig. 18C is a graph illustrating Ad-I NSM 1 p-/Wef vector expressed Metridia luciferase expression specifically in Insulinoma β " Ι -1 , MIN, RIN, and pancreatic epithelioid carcinoma PANC-1 cells were used. Values are expressed as ratios between extracellular and intracellular luciferase activity. *: p<0.05, **: p<0.01 , ***: p<0.001 (n=3).

Fig. 19 is a graph illustrating Metridia luciferase secretion in xenograft human tumor culture. Ad-I NSM 1 p-/Wef vector specifically expressed Metridia luciferase in ex vivo NE tumor cells. Cells harvested from established xenograft human tumors were infected with Ad-I NSM 1 p-Met (100 MOI) and AdSV40-/_i/c2 (100 MOI). Data points are displayed as ratios between extracellular and intracellular luciferase luminescence. *: p<0.05, **: p<0.01 , ***: p<0.001 (n=4).

Figs. 20A-20C illustrate Ad-I NSM 1 p-Gau vector expressed Gaussia luciferase specifically in INSM1-positive cell lines. An increasing Ad-INSM1 p-Gau concentration (0-50 MOI) and a constant AdSV40-Luc2 concentration (5 MOI) was used to infect INSM1-positive (solid lines) and -negative (dot lines) cell lines in culture.

Fig. 20A is a graph illustrating Ad-INSM1 p-Gau vector expressed Gaussia luciferase specifically in NE lung cancer H1155, UMC-11 , H82, and lung adenosquamous carcinoma H596 cells. Fig. 20B is a graph illustrating Ad-INSM1 p-Gau vector expressed Gaussia luciferase specifically in Neuroblastoma SK-N-Be(2) and SH-SY5Y, medulloblastoma D283Med, and cervical adenocarcinoma HeLa cells.

Fig. 20C is a graph illustrating Ad-INSM 1 p-Gau vector expressed Gaussia luciferase specifically in Insulinoma βΤΟ-1 , MIN, RIN, and pancreatic epithelioid carcinoma PANC-1 cells were used. Values are expressed as ratios between extracellular and intracellular luciferase activity. *: p<0.05, **: pO.01 , ***: pO.001 (n=3).

Fig. 21 is a graph illustrating oncolytic virus in combination with Ad-INSM1 p-Gau increases sensitivity of Gaussia luciferase detection. The combination of Ad-INSM1 p-Gau and Ad-INSM1 p-A24E1A was able to increase the sensitivity of luciferase detection after 6 days post infection. To test the effects of Ad-INSM1 p-Gau in combination with the conditionally replicating adenovirus Ad-INSM1 p-A24E1A, cells were infected with combination of 10 MOI Ad-INSM1 p-Gau and Ad-INSM1 p-A24E1A (a total of 20 MOI). Media were collected 2, 4, and 6 days after infection and showed the highest luciferase activity at day 6. *: p<0.05, **: p<0.01 (n=3).

Fig. 22 is a graph illustrating in vivo Gaussia secretion assay in a mouse xenograft tumor model. The combination of Ad-INSM1 p-Gau and Ad-INSM 1 p-A24E1A was able to express detectable Gaussia luciferase in circulation 12 days after infection in vivo. A mixture of Ad-INSM1 p-Gau in combination with the Ad-INSM1 p-A24E1A conditionally replicating adenovirus (1 x 109 ifu total) was injected into established xenograft tumors (H1155 NE lung cancer cells). Serum was collected from each animal (n=3) 3 to 12 days post infection. **: p<0.01 (n=3).

Fig. 23 is a series of digital images illustrating Western blot analyses of viral protein expression. To examine the expression of viral proteins, Ad-INSM1-HSVTK and Ad-INSM 1- E1A-HSVTK were used to infect H155 (INSM 1+ LCLC), H69, H82, UMC1 1 (all INSM1 +

SCLC), and H596 (INSM1- non-SCLC) cells at 50 MOI. After incubation for 48 hours, total protein extracts were isolated from each cell line. The viral combination expressed higher levels of HSVTK than either virus alone.

Fig. 24 is a graph illustrating viral replication in INSM1 positive SCLC cells. H1155 cells were plated on 6-well plates at a density of 2* 10 6 cells/well and infected with a viral concentration of 10 MOI or 200 MOI. After incubation for approximately 3 days, cells were collected and viral particles released by freeze thaw lysis. The isolated virus were then titered by immunostaining. (n=6)

Fig. 25 is a series of graphs illustrating cell survival assay. An MTS colorimetric assay was conducted with 5 lung cancer cell lines: H155 (INSM1 + LCLC), H69, H82, UMC11 (all INSM1 + SCLC), and H596 (INSMT non-SCLC). Cells are first treated with virus ranging from 0-200 MOI. Three days after infection with viral constructs, cells were treated with 100 μΜ GCV for 5 days. Cell survival was measured by MTS colorimetric analysis at 490 nm. (n=3)

Fig. 26 is a series of graphs illustrating cell survival assay. An MTS assay was conducted with 5 lung cancer cell lines: H155 (INSM1 + LCLC), H69, H82, UMC11 (all INSM1 + SCLC), and H596 (INSM1 - non-SCLC). Cancer cells were infected with 50 MOI viruses for 3 days, after which 0-200 μΜ ganciclovir was added for 5 additional days. Cell survival was measured by MTS colorimetric analysis at 490 nm. (n = 3).

Figs. 27A-27C illustrate subcutaneous xenograft tumors. A total of 10 9 cells were injected subcutaneously into the right flank of 4 week old Nu/Nu mice. After establishment of tumor, the viral combination was introduced via injection into the tumor. GCV treatment was started 3 days after infection to allow for viral replication to occur. In certain tumors, a second viral boost was introduced after tumor growth accelerated despite viral treatment. Tumors were measured daily in 3 dimensions to determine volume.

Fig. 27A is a graph illustrating H1155 large cell lung carcinoma.

Fig. 27B is a graph illustrating UMC11 SCLC.

Fig. 27C is a graph illustrating H82 SCLC.

Fig. 28 are digital images illustrating subcutaneous xenograft tumors. Photograph of H1 155 subcutaneous tumors after completion of treatment with combination virus (left) or control Ad-INSM1-Luc2 virus (right).

Fig. 29 is a graph illustrating nasal inhalation delivers viruses primarily into the left and right lungs. Nasal inhalation of virus was conducted using 10 9 ifu Ad-CMV-Luc2 at a total volume of 40 into 4 week old Nu/Nu mice. After incubation for 24 h post-inhalation, organs were harvested from each mouse the protein extracts were produced. Luciferase activity was detected in each organ using 20 of extract to be used for the Pierce

Luciferase Glow Assay Kit. (n=3).

Fig. 30 are digital images illustrating orthotopic lung tumor generation. H1155 tumor cells were injected into the left lung of Nu/Nu mice. A total of 10 9 cells were injected mixed with a 1 :5 concentration of matrigel. After 3 days, 10 9 ifu Ad-INSM1-Luc2 was introduced to the tumor bearing mouse by nasal inhalation. Luciferin substrate was injected

intraperitoneally at day 4 to detect luciferase activity. This process was repeated for days 8 and 12, after which the mouse was sacrificed and the lung removed (right).

Fig. 31 are digital images illustrating treatment and monitoring of orthotopic lung tumors. UMC11 SCLC tumor cells were first infected with 10 MOI Ad-INSM1-Luc2 for 24h. After infection, 10 9 cells were mixed with a 1 :5 concentration of matrigel and injected into the left lung of Nu/Nu mice. After 3 days, 10 9 ifu combination virus or Ad-INSM1-Luc2 was introduced to the tumor bearing mice by nasal inhalation of 40 μί of viral solution. Luciferin substrate was IP injected every 5 days to detect luciferase activity. Fig. 32: Nucleotide sequence of the 2Xins-INSM1 p--2XNRSE-A24E1A-IRES-HSV-tk construct that is inserted into the adenovirus 5 (Ad5) viral vector (SEQ ID Nos: 11).

Fig. 33: Nucleotide sequence of the 2Xins-INSM1 p--2XNRSE-A24E1A-IRES-HSV-tk construct that is inserted into the adenovirus 5 (Ad5) viral vector (SEQ ID Nos: 12).

Fig. 34: Nucleotide sequence of a 2Xins-INSM1 p-2XNRSE-A24E1A-3'-untranslated region construct that may be inserted into an adenovirus 5 (Ad5) viral vector (SEQ ID NO: 13).

Fig. 35: Nucleotide sequence of a 2Xins-INSM 1 p-2XNRSE-Gaussia-3'-untranslated region construct that may be inserted into an adenovirus 5 (Ad5) viral vector (SEQ ID NO: 14).

DETAILED DESCRIPTION

Before the present disclosure is described in greater detail, it is to be understood that this disclosure is not limited to particular embodiments described, and as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present disclosure will be limited only by the appended claims.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure, the preferred methods and materials are now described.

All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the

publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present disclosure is not entitled to antedate such publication by virtue of prior disclosure. Further, the dates of publication provided could be different from the actual publication dates that may need to be

independently confirmed.

As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present disclosure. Any recited method can be carried out in the order of events recited or in any other order that is logically possible.

Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of medicine, organic chemistry, biochemistry, molecular biology, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature.

It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a support" includes a plurality of supports. In this specification and in the claims that follow, reference will be made to a number of terms that shall be defined to have the following meanings unless a contrary intention is apparent.

As used herein, the following terms have the meanings ascribed to them unless specified otherwise. In this disclosure, "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. patent law and can mean " includes," "including," and the like; "consisting essentially of" or "consists essentially" or the like, when applied to methods and compositions encompassed by the present disclosure refers to compositions like those disclosed herein, but which may contain additional structural groups, composition components or method steps (or analogs or derivatives thereof as discussed above). Such additional structural groups, composition components or method steps, etc., however, do not materially affect the basic and novel characteristic(s) of the compositions or methods, compared to those of the corresponding compositions or methods disclosed herein.

Prior to describing the various embodiments, the following definitions are provided and should be used unless otherwise indicated.

Abbreviations

NE, neuroendocrine; INSM1 ,insulinoma-associated-1 ; NRSEs, neuronal restrictive silencerelements;HSV-tk,herpessimplexvirusthymidinekinase;GC V, ganciclovir; DMEM, DulbeccoModifiedEagleMedium;MOI,multiplicitiesofinfection;CA R,coxsackievirusand adenovirus receptor; Luc2, Iuciferase2; Gau, Gaussia, Met, Metridia; RFU, reflective fluorescence unit. Definitions

As used in the specification and in the claims, the term "construct" or "expression construct" refers to a functional DNA nucleotide sequence that is artificially constructed to transfer or express one or more genes of interest.

The terms "neuroendocrine cancer" or "neuroendocrine tumor" as used herein refer to a cancer that arises from the neuroendocrine system, a diffuse system in which the nervous system and the hormones of the endocrine glands interact, or from non-endocrine cells by acquiring some of the properties of neuroendocrine cells through an oncogenic process such as Selective Tumour gene Expression of Peptides essential for Survival (STEPS) (see, North (2000) Exper. Physiol. 85S:27S-40S). Most of the well-described adult neuroendocrine tumors are distinctive and arise from a known primary site, including the carcinoid, pheochromocytoma, and Merkel's cell tumors. Carcinoid tumors can be benign or malignant. Carcinoid cancers include stomach, pancreas, colon, liver, lung, ovarian, breast, testicular, and cervical cancer. Neuroendocrine tumors of the lungs are classified as small cell carcinoma. It is characterized by its origin in large central airways and histological composition of sheets of small cells with scanty cytoplasm. Small cell carcinoma is very aggressive, metastazing early and often. Pheochromocytoma is a cancer of the adrenal medulla. This condition often causes the adrenal glands to make too much catecholamine. Pheochromocytoma may arise as part of a condition called multiple endocrine neoplasia (MEN) syndrome, which can result in other cancers of the endocrine system and hormonal abnormalities. Merkel's cell tumors are cancers that form on or just beneath the skin, but sometimes are also thought to arise from underlying soft tissue. They are also known as neuroendocrine cancer of the skin. Merkel's cell tumors are fast-growing and often spread to other parts of the body. In particular embodiments of the present invention, a

neuroendocrine cancer is a carcinoid cancer such as breast cancer or SCLC.

The term "neuronal restrictive silencer element" or "NRSE" as used herein refers to a DNA segment that is known to mediate transcriptional repression of many neuron-specific genes via the neuron-restrictive silencer factor (NRSF) or repressor element silencing transcription factor. The neuron-restrictive silencer element (NRSE) has been identified in several neuronal genes and confers neuron specificity by silencing transcription in non- neuronal cells. NRSE elements when bound by its cognate protein, the neuron restrictive silencing factor, NRSF, can strongly repress transcription in non-neuronal cells as well as allow transcription of the same gene in neuronal cells. Examples of neuronal restrictive silencer elements (NRSEs) include those derived either from the mouse nicotinic

acetylcholine receptor (nAChR) or the rat superior cervical ganglion 10 (SCG10) promoters. Multiple neuronal genes have been shown to be repressed by NRSF protein via a NRSE element located in their promoter regions. Other neuronal genes repressed by NRSF in non-neuronal cells include protocadherin, tryptophan hydroxylase-2, mu opioid receptor, tyrosine hydroxylase, N-methyl-D-aspartate receptor 2B, proprotein convertase 2, glutamate receptor 2, GluR2, arginine vasopressin, brain-derived neutrophic factor, neural-specific type II sodium channel, and dopamine beta hydroxylase genes. (See also, U.S. Patent

Application Publication No. 2006/0121013)

The term "insulator element" as used herein refers to a DNA segment that has the ability to protect genes from inappropriate signals originating from the surrounding environment by acting as a physical barrier or boundary. An insulator element blocks the interaction between a promoter and enhancers when it is inserted between them, or it confers expression of integrated foreign genes independent of their position in the chromatin. The 5' HS4 element, derived from the chicken .beta.-globin locus (the first insulator identified in vertebrates), has been used with success to improve heterologous construct expression in transgenic animals. The chicken β-globin HS4 insulator element has been shown to block the actions of enhancer elements in addition to functioning as a physical boundary that can prevent the spread of gene silencing (14-20). In this

embodiment of the construct, the insulator element is used to prevent the adenoviral sequences from potentially interrupting the INSM1 promoter activity and to prevent the interference from the viral backbone with respect to the tissue selectivity of the promoter incorporated into the viral vectors.

The term "Internal Ribosome Entry Site (IRES)" as used herein refers to an RNA element that allows for translation initiation in an end-independent manner. In eukaryotic translation, initiation typically occurs at the 5' end of mRNA molecules, since 5' cap recognition is required for the assembly of the initiation complex. The location for IRES elements is often in the 5'UTR, but can also occur elsewhere in mRNAs and may be used to create multigene, or polycistronic, messages. IRES elements are able to bypass the ribosome scanning model of 5' methylated Cap dependent translation and begin translation at internal sites (Pelletier and Sonenberg, 1988). IRES elements from two members of the picornavirus family (polio and encephalomyocarditis) have been described (Pelletier and Sonenberg, 1988), as well an IRES from a mammalian message (Macejak and Sarnow, 1991). IRES elements can be linked to heterologous open reading frames. Multiple open reading frames can be transcribed together, each separated by an IRES, creating polycistronic messages. By virtue of the IRES element, each open reading frame is accessible to ribosomes for efficient translation. Multiple genes can be efficiently expressed using a single promoter/enhancer to transcribe a single message (see U.S. Pat. Nos.

5,925,565 and 5,935,819, herein incorporated by reference).

The term "ganciclovir" as used herein refers to 2-amino-9-{[(1 ,3-dihydroxypropan-2- yl)oxy]methyl}-6,9-dihydro-3/-/-purin-6-one, a nucleoside analog. The term "nucleoside analog" as used herein refers to a compound having a molecular structure similar to a nucleoside (e.g., adenosine, guanosine, thymidine and cytidine, and deoxy derivatives thereof) and/or a nucleic base (e.g., adenine, guanine, thymine and cytosine). The term "analog" indicates that the compound is capable of interacting (e.g., as a substrate and/or inhibitor) with an enzyme for which a nucleoside and/or nucleic base is a natural substrate and/or inhibitor.

In embodiments of the disclosure, the nucleoside analog can an antiviral activity which is triggered by thymidine kinase (TK) and results in a selective nucleoside analog. The nucleoside analog is converted into an active, phosphorylated form in cells having the TK (i.e., cells infected with an expression vector encoding and expressing the TK) while remaining in the original inactive, non-phosphorylated form in other cells. By remaining in an inactive form in non-infected cells, side effects caused by damage to non-infected cells are reduced considerably.

The term "reporter gene" as used herein refers to a gene, usually a foreign or modified gene, that is added to a construct and is expressed due to the promoter in the construct and the expression allows easy identification of cells or tissues that have taken up the construct. Common reporter genes include the gene that encodes jellyfish green fluorescent protein, which causes cells that express it to glow green under UV light, and the firefly luciferase gene which causes light emission when its substrate luciferin is added. Reporter genes are often placed downstream of the promoter region and in the proximity of the gene of interest to ensure that they are expressed together and not separated by crossover events.

The term "toxin gene" s used herein refers to a gene that encodes a toxin that is capable of being readily produced either under the regulatory control of the INSM1 promoter. A "toxin" is a gene product(s) that causes or leads to the destruction or incapacitation of a cell. This definition is intended to include the induction of indigenous events leading to cell death, such as apoptosis or necrosis. A "toxin" may, for example, be a compound that induces conditional lethality, i.e., cell death requires both expression of a conditional toxin gene (for example, thymidine-kinase) and the exogenous administration of a compound (for example, ganciclovir or acyclovir) that together produce a lethal effect. Another example is the combination of the gene encoding cytosine deaminase and the pro-drug 5- fluorocytosine. For example, a suitable toxin may be one of the many toxic peptides known in the art. In addition, the toxin should be capable of killing tumor cells or, optionally, the toxin may also kill neighboring cells, a "bystander" effect, but it should not have substantial systemic effects. There are numerous toxins from plants, animals, and bacteria satisfying these criteria, including naturally occurring, modified and synthetic toxins. Examples of toxins include without limitation synthetic and natural lytic peptides, cholera toxin, diphtheria toxin, Pseudomonas toxin, ricin toxin, cecropins, defensins, sarcotoxins, melittins, and magainins. One suicide gene therapy uses the gene herpes simplex virus thymidine kinase and ganciclovir. The disadvantage to this system includes significant liver toxicity unless precaution is taken to decrease its expression in liver cells. (See also, discussion in

PCT/USOO/0633 published as WO005377; U.S. Pat. Nos. 5,789,542 and 6,566,334.)

The term "viral vector" as used herein refers to a virus that is competent to infect a mammalian host cell and can be used to deliver the construct to the target cells or tumor or to an animal systemically. One example of a viral vector is the first generation E1/E3 deleted non-replicating Ad5 vector, but other forms of viral delivery systems are known and could be used. One of the disadvantages of the non-replicating adenovirus is the lack of persistence in vivo and one embodiment could be the use of a conditionally replicating oncolytic adenovirus. Additional examples of viral delivery systems include viruses that would result in more permanent expression such as lentivirus or adeno-associated virus (AAV). The advantage to these two viral systems is that they can be manipulated to alter their tropism for different cell types making them a more flexible platform.

Neuroendocrine tumors that can be treated or diagnosed using the described construct include without limitation retinoblastoma, medullablastoma, neuroblastoma, small cell lung carcinoma, non-small cell carcinoma with neuroendocrine phenotype, carcinoid, insulinoma, pheochromocytoma, medullary thyroid carcinoma, pituitary tumors, prostate carcinoma, and retinoblastoma tumors.

In describing and claiming the disclosed subject matter, the following terminology will be used in accordance with the definitions set forth below.

Description

Insulinomas are the most common type of islet cell tumors. Characteristically, patients with insulinomas can develop complications associated with hyperinsulinemia.

Modification of an INSM1 promoter region to incorporate DNA elements that have silenced expression of neuronal genes in non-neuronal cells has increased the effectiveness and safety of using the INSM1 promoter for tumor treatment. To increase the safety of the transcriptionally-regulated suicide gene therapy, various DNA elements were included in the 1.7 kilobase pair INSM1 promoter for reduction in expression in unwanted tissues. The first modification was addition of two tandem copies of neuronal restrictive silencer elements (NRSEs) derived either from the mouse nicotinic acetylcholine receptor (nAChR) or the rat superior cervical ganglion 10 (SCG10) promoters. These NRSEs were placed in the construct directly downstream of the INSM 1 promoter sequence. The most effective construct was the nAChR NRSE element positioned downstream of the INSM1 promoter. This construct increased the tissue specificity of the INSM1 promoter without a significant decrease in its activity. The INSM1 promoter has been linked to a toxin for tumor therapy and placed the constructs into the adenovirus 5 viral vector. Constructs were also tested with an insulator element with the INSM1 promoter to decrease the interference of the viral genome on its expression. A construct using the chicken HS4p-globin insulator element was shown to work as expected. Linking the construct to a reporter gene allowed for the detection of the placement of the viral vector, and can be used for diagnosing

neuroendocrine tumors. Further constructs that do not decrease the INSM 1 promoter activity but significantly augment the tumor specificity of the promoter have also been generated and can be used for treatment and diagnosing of neuroendocrine tumors.

The present disclosure encompasses embodiments of a modified neuroendocrine tumor-specific adenovirus expression vector that expresses a mutated adenovirus E1A gene (Δ24Ε1Α) under the regulatory control of an INSM1 (insulinoma-associated-1) promoter. In addition, an IRES element is included to allow the expression of at least one second open reading frame encoding a polypeptide. This second polypeptide can be any that is useful for the detection of a neuroendocrine tumor cell, or for the reducing the proliferation or viability of the cell. For example, but not intended to be limiting the second polypeptide can be a tumor cell-specific lethal polypeptide or a conditionally lethal polypeptide such as thymidine kinase.

The expression vectors of the disclosure, therefore, enable selective gene expression specifically in neuroendocrine tumor cells, and allow for the expression of a heterologous ORF. The heterologous gene can be either a full genomic sequence (e.g., including introns), synthetic nucleic acid or a cDNA copy of a gene of interest, which encodes a protein or a polypeptide of interest, wherein the polypeptide includes biologically active ("bioactive") protein fragments. In a preferred embodiment, cDNA sequences are used for the purposes of the present technology due to the reduction in genomic complexity provided by removal of mRNA splice sites.

The ORF sequence may be selected from the group of reporter genes, cytotoxic tumor suppressor genes, toxin genes, prodrug activating genes and proapoptotic genes. In some embodiments, the second ORF sequence can be a reporter gene. As the name implies, a reporter gene does not confer any selective advantage on the cell into which it is introduced. Rather, a reporter gene encodes a product that confers on the cell a detectable biochemical or visually observable (e.g., fluorescent) phenotype. The reporter polypeptide can also include a fused or hybrid polypeptide in which another polypeptide is fused at the N-terminus or the C-terminus of the polypeptide or fragment thereof. A fused polypeptide is produced by cloning a nucleic acid sequence (or a portion thereof) encoding one polypeptide in-frame with a nucleic acid sequence (or a portion thereof) encoding another polypeptide. Techniques for producing fusion polypeptides are known in the art, and include ligating the coding sequences encoding the polypeptides so that they are in-frame and translation of the fused polypeptide is under the control of the TR cassette.

One commonly used class of reporter genes encodes an enzyme or other biochemical marker, which, when expressed in a mammalian cell, cause a visible change in the cell or the cell environment. Such a change can be observed directly, or can involve the addition of an appropriate substrate that is converted into a detectable product or the addition and binding of a metabolic tracer. Examples of these reporter genes are the bacterial lacZ gene which encodes the b-galactosidase (b-gal) enzyme, Firefly luciferase (Coleoptera beetle), Renilla luciferase (sea pansy), Herpes Simplex 1 thymidine kinase (HSV1-TK) and the mutant Herpes Simplex 1 thymidine kinase (HSV1-sr39tk) genes. In the case of b-gal, incubation of expressing cells with halogen-derivatized galactose results in a colored or fluorescent product that can be detected and quantitated histochemically or fluorimetrically. Other useful reporter genes encode proteins that are naturally fluorescent, including the (green fluorescent protein (GFP), enhanced yellow fluorescent protein (EYFP), or monomeric red fluorescent protein (mRFP1).

The second ORF sequence can encode a cytotoxic tumor suppressor gene that encodes a polypeptide capable of suppressing the neoplastic phenotype and/or inducing apoptosis. Examples of tumor suppressor genes useful in the practice of the present technology include the p53, adenomatous polyposis coli (APC), Wilm's Tumor (WT-1), retinoblastoma gene (Rb), Neurofibromatosis-1 (NF-1), NF-2 and von Hippel-Lindau (VHL) genes. In a preferred embodiment, the cytotoxic tumor suppressor gene is the p53 gene.

In one embodiment, the second ORF sequence may encode a "toxin gene" that binds to cellular receptor proteins and after uptake interferes with protein synthesis by blocking ribosome assembly or function. Examples of toxin genes include proteins such as Pseudomonas exotoxin (e.g., Exotoxin A or "ETA"), ricin toxin, diphtheria toxin, and the like. In one embodiment, the toxin gene is the diphtheria toxin gene.

In other embodiments, the second ORF sequence is a prodrug activating gene (e.g., drug-susceptibility or suicide gene) that codes for a protein that converts a prodrug that lacks a therapeutic effect into a drug which renders a cell expressing said gene susceptible to death following exposure to said prodrug. Examples of pro-drug genes include the thymidine kinase of Herpes Simplex Virus (HSV-tk), cytochrome P450, human deoxycytidine kinase and the bacterial enzymes cytosine deaminase and guanine phosphoribosyl transferase (gpt) genes. Cells that express these genes are rendered sensitive to the prodrugs ganciclovir (HSV-tk), cyclophosphamide (cytochrome P450), cytosine arabinoside (deoxycytidine kinase), 5-fluorocytosine (bacterial cytosine deaminase) or thioxanthine (gpt). In a preferred embodiment, the prodrug activating gene is the HSV-tk gene which can also provide an important therapeutic advantage. During TK catalysis of the antiviral guanosine analogue ganciclovir, apoptotic molecules are released that kill surrounding cells by a process termed "bystander" killing. A limited number of target cells may initially express the HSV-tk gene, this localized cytocidal effect provides a therapeutic effect to adjacent non- expressing, undesired bystander cells.

It has been found that the expression level of the second polypeptide expressed from the IRES can be lower than expected. However, co-infection of a recipient cell with an expression vector expressing the second open-reading frame from another promoter that is not an IRES results in synergistic expression of the second open-reading frame.

Accordingly, one aspect of the disclosure encompasses embodiments of a method of modulating the proliferation or viability of a neuroendocrine tumor by co-infecting the target tumor

To increase the treatment options for insulinoma patients, a conditionally replicating adenovirus has been generated that can specifically replicate and express therapeutic genes in neuroendocrine (NE) tumors. The promoter-specific expression of these adenoviruses is regulated upstream by an INSM1 (insulinoma-associated-1) promoter that is silent in normal adult tissues but active in developing NE cells and NE tumors. By placing the INSM1- promoter with an insulator (HS4) and two copies of neuronal restrictive silencer elements (NRSEs) in an adenoviral vector, the construct can retain tumor specificity and drive expression of a mutated adenovirus E1A gene (Δ24Ε1Α) and the herpes simplex virus thymidine kinase (HSV-tk) gene. Data obtained using pancreatic cell lines in vitro and a subcutaneous mouse tumor model revealed that the I NSM1 -promoter driven viruses were able to replicate specifically in the I NSM1 -positive cells. I NSM 1 -specific HSV-tk expression in combination with ganciclovir treatment displayed dose-dependent tumor cell killing in insulinomas, leaving I NSM1 -negative cells unharmed. When the I NSM1 -promoter driven HSV-tk was combined with Δ24Ε1Α and I NSM1 p- HSV-tk (K5) viruses, the co-infected insulinoma expressed higher levels of HSV-tk and more efficient tumor suppression as compared to the I NSM1 p- HSV-tk virus alone. Taken together, INSM I p-driven conditionally replicating adenoviruses represent a new tool for the treatment of insulinoma that provides clinician additional options to combat this disease.

Accurate detection of neuroendocrine (NE) tumors is critically important for better prognosis and treatment outcomes in patients. To demonstrate the efficacy of using an adenoviral vector for the detection of NE tumors, a pair of adenoviral vectors was

constructed that in combination can conditionally replicate and release Gaussia luciferase into the circulation after infecting the NE tumors. The expression of these two vectors is regulated upstream by an I NSM1 -promoter (insulinoma-associated-1) that is specifically active in NE tumors and developing NE tissues, but silenced in normal adult tissues. To retain the tumor-specificity of the INSM1 promoter, the promoter was modified using the core insulator sequence from the chicken β-globin HS4 insulator and the neuronal restrictive silencing element (NRSE). This modified I NSM1 -promoter can retain NE tumor specificity in an adenoviral construct while driving a mutated adenovirus E1A gene (Δ24Ε1Α), the

Metridia, or Gaussia luciferase gene.

The in vitro cell line and mouse xenograft human tumor studies revealed the NE specificity of the I NSM1 -promoter in NE lung cancer, neuroblastoma, medulloblastoma, retinoblastoma, and insulinoma. When the I NSM1 -promoter driven Gaussia luciferase was combined with Δ24Ε1Α, the co-infected NE tumor secreted higher levels of Gaussia luciferase as compared to the INSM1 p-Gai/ss/ ' a virus alone. In a mouse subcutaneous xenograft tumor model, the combination viruses secreted detectable level of Gaussia luciferase after infecting an I NSM 1 -positive NE lung tumor for at least 12 days. Therefore, the I NSM1 -promoter specific conditional replicating adenovirus represents a sensitive diagnostic tool to aid clinicians in the detection of NE tumors.

One aspect of the disclosure, therefore, encompasses embodiments of a

neuroendocrine tumor-specific viral expression vector whose genome comprises: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

In some embodiments of this aspect of the disclosure, the open reading frame nucleotide sequence operably linked to the IRES element can encode the conditionally lethal toxin herpes simplex virus thymidine kinase ( SV-tk).

In some embodiments of this aspect of the disclosure, the secreted bioluminescent reporter molecule can be a luciferase 2, a Renilla luciferase, a Metridia luciferase, or a Gaussia luciferase.

In some embodiments of this aspect of the disclosure, the viral expression vector can be a non-replicating Ad5 adenoviral vector.

In some embodiments of this aspect of the disclosure, the human INSM1 promoter comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region is a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements comprises a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter has at least 95% similarity to SEQ ID NO: 7 and encodes the tumor-selective modified E1A polypeptide Δ24Ε1Α, the IRES element has the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'-untranslated region has a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

In some embodiments of this aspect of the disclosure, the nucleotide sequence operably linked to the IRES element can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encoding the herpes simplex virus thymidine kinase ( SV-tk).

In some embodiments of this aspect of the disclosure, the viral expression genome can comprise the nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 1.

Another aspect of the disclosure encompasses embodiments of a composition comprising a first neuroendocrine tumor-specific viral expression vector whose genome comprises: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

In some embodiments of this aspect of the disclosure, the nucleotide sequence operably linked to the IRES element can encode the conditionally lethal toxin herpes simplex virus thymidine kinase ( SV-tk).

In some embodiments of this aspect of the disclosure, the secreted bioluminescent reporter molecule can be a luciferase 2, a Renilla luciferase, a Metridia luciferase, or a Gaussia luciferase.

In some embodiments of this aspect of the disclosure, the viral expression vector can be a non-replicating Ad5 adenoviral vector.

In some embodiments of this aspect of the disclosure, the human INSM1 promoter can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region can be a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter can have at least 95% similarity to SEQ ID NO: 7 and encode the tumor-selective modified E1A polypeptide Δ24Ε1Α, an IRES element can have the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'-untranslated region can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

In some embodiments of this aspect of the disclosure, the composition can further comprise the herpes simplex virus thymidine kinase ( SV-tk) having the nucleotide sequence having at least 95% similarity to SEQ ID NO: 9.

In some embodiments of this aspect of the disclosure, the viral expression genome can comprise the nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 1.

In some embodiments of this aspect of the disclosure, the composition can further comprise a pharmaceutically acceptable carrier.

In some embodiments of this aspect of the disclosure, the composition can further comprise a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and (d) a nucleotide sequence encoding a 3'-untranslated region, wherein: (i) said second viral expression vector is competent to infect at least some mammalian cells; (ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences; (iii) said INSM 1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells; wherein the level of expression of the polypeptide toxin or the polypeptide reporter molecule from the combination of the first and second vectors in a recipient cell is synergistically greater compared to expression from the first or second vector alone.

In some embodiments of this aspect of the disclosure, the first and the second viral expression vectors can each express the conditionally lethal toxin herpes simplex virus thymidine kinase (HSV-tk).

In some embodiments of this aspect of the disclosure, the first and the second viral expression vectors can each express a reporter molecule.

In some embodiments of this aspect of the disclosure, the first and the second viral expression vectors are each genetically modified non-replicating Ad5 adenoviral vectors.

In some embodiments of this aspect of the disclosure, the composition can be formulated for directed delivery to a tumor, intravenous delivery to a tumor, or respiratory delivery to a lung tumor.

Yet another aspect of the disclosure encompasses embodiments of a method of increasing the tumor-specific expression of a polypeptide from a viral vector, said method comprising delivering to a neuroendocrine tumor cell or tumor a first neuroendocrine tumor- specific viral expression vector whose genome comprises: (a) a human Insulinoma- associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a tumor-selective modified human E1A polypeptide lacking the amino acid residues 121-128 of the amino acid sequence SEQ ID NO: 8; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements (NRSE) downstream from said promoter; (d) an IRES element operatively linked upstream of one or more open reading frame nucleotide sequences encoding at least one of a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a secreted bioluminescent reporter molecule; and (e) at least one nucleotide sequence encoding a 3'- untranslated region wherein: (i) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of the INSM1 promoter or the influence of the neuronal restrictive silencer elements on the transcription of said one or more polypeptide encoding nucleotide sequences; (ii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iii) said pair of tandem neuronal restrictive silencer elements are operatively linked to the INSM1 promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells.

In some embodiments of this aspect of the disclosure, the method can further comprise delivering to the neuroendocrine tumor cell or tumor a second neuroendocrine tumor-specific viral expression vector, the genome of said second viral expression vector comprising: (a) a human Insulinoma-associated 1 promoter (INSMI p) operatively linked to a nucleotide sequence encoding a polypeptide toxin lethal or conditionally lethal to cells in which the toxin is expressed and a polypeptide reporter molecule; (b) an insulator region upstream from said promoter, wherein said insulator region comprises an insulator element or a tandem pair of core insulator elements; (c) a pair of tandem neuronal restrictive silencer elements downstream from said promoter; and (d) a nucleotide sequence encoding a 3'- untranslated region, wherein: (i) said second viral expression vector is competent to infect at least some mammalian cells; (ii) said insulator region is adapted to block the influence of viral regulatory elements on the transcription within mammalian cells of said one or more nucleotide sequences, but not to affect the influence of said promoter or the influence of said neuronal restrictive silencer elements on the transcription of said one or more nucleotide sequences; (iii) said INSM1 promoter is adapted to cause the selective transcription of said one or more nucleotide sequences in tumor cells of neuroendocrine origin; and (iv) said tandem neuronal restrictive silencer elements are operatively linked to said promoter, and are adapted to selectively repress the transcription of said one or more nucleotide sequences in non-neuronal cells; wherein the level of expression of the at least one of the polypeptide toxin and the polypeptide reporter molecule from the combination of the first and second vectors in the recipient cell or tumor is synergistically greater compared to expression from the first or second vector alone.

In some embodiments of this aspect of the disclosure, the first and second viral vectors can be delivered to the recipient cell or tumor sequentially.

In some embodiments of this aspect of the disclosure, the first and second viral vectors can be co-delivered in a single pharmaceutically acceptable composition.

In some embodiments of this aspect of the disclosure, the first neuroendocrine tumor-specific viral expression vector the human INSM1 promoter can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region can be a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each of said core elements comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter can have at least 95% similarity to SEQ ID NO: 7 and encode the tumor-selective modified E1A polypeptide Δ24Ε1Α, the IRES element can have the nucleotide sequence having at least 95% similarity to SEQ ID NO: 6; and the 3'-untranslated region can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

In some embodiments of this aspect of the disclosure, the nucleotide sequence operably linked to the IRES element can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encodes the herpes simplex virus thymidine kinase ( SV-tk).

In some embodiments of this aspect of the disclosure, the first neuroendocrine tumor-specific viral expression genome can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 1.

In some embodiments of this aspect of the disclosure, the second neuroendocrine tumor-specific viral expression vector the human INSM 1 promoter can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 1 , the insulator region can be a chicken HS4 β-globin insulator element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 3 or a pair of tandem core chicken HS4 β-globin insulator elements, each core element comprising a nucleotide sequence having at least 95% similarity to SEQ ID NO: 4, the pair of tandem neuronal restrictive silencer elements can comprise a nucleotide sequence having at least 95% similarity to SEQ ID NO: 2, the nucleotide sequence operably linked to the INSM1 promoter can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 9 and encode the herpes simplex virus thymidine kinase ( SV-tk), and the 3'- untranslated region can have a nucleotide sequence having at least 95% similarity to SEQ ID NO: 10.

In some embodiments of this aspect of the disclosure, at least one of the viral expression vectors thereof can encode a reporter molecule, and the method further comprises observing subsequent expression of the encoded reporter molecule in the patient's tissues as a measure of the presence or the extent of a neuroendocrine tumor. In some embodiments of this aspect of the disclosure, the reporter molecule can be Gaussia luciferase and a nucleotide sequence encoding said luciferase has at least 95% similarity to SEQ ID NO: 5.

In some embodiments of this aspect of the disclosure, the first and second neuroendocrine tumor-specific viral expression vectors can each comprise a nucleotide sequence encoding a polypeptide toxin and wherein, if said polypeptide toxin is only conditionally lethal, then said method additionally can comprise the step of providing conditions that produce the lethal phenotype; whereby cells of the neuroendocrine tumor are selectively killed.

In some embodiments of this aspect of the disclosure, the method can further comprise contacting the neuroendocrine cell with ganciclovir thereby selectively killing cells of the neuroendocrine tumor.

It should be emphasized that the embodiments of the present disclosure, particularly any "preferred" embodiments, are merely possible examples of the implementations, merely set forth for a clear understanding of the principles of the disclosure. Many variations and modifications may be made to the above-described embodiment(s) of the disclosure without departing substantially from the spirit and principles of the disclosure. All such modifications and variations are intended to be included herein within the scope of this disclosure, and protected by the following claims.

The specific examples below are to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. Without further elaboration, it is believed that one skilled in the art can, based on the description herein, utilize the present disclosure to its fullest extent. All publications recited herein are hereby incorporated by reference in their entirety.

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to perform the methods and use the compositions and compounds disclosed and claimed herein. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in °C, and pressure is at or near atmospheric. Standard temperature and pressure are defined as 20 °C and 1 atmosphere.

It should be noted that ratios, concentrations, amounts, and other numerical data may be expressed herein in a range format. It is to be understood that such a range format is used for convenience and brevity, and thus, should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. To illustrate, a concentration range of "about 0.1 % to about 5%" should be interpreted to include not only the explicitly recited concentration of about 0.1 wt% to about 5 wt%, but also include individual concentrations (e.g., 1 %, 2%, 3%, and 4%) and the sub-ranges (e.g., 0.5%, 1.1 %, 2.2%, 3.3%, and 4.4%) within the indicated range. The term "about" can include ±1 %, ±2%, ±3%, ±4%, ±5%, ±6%, ±7%, ±8%, ±9%, or ±10%, or more of the numerical value(s) being modified.

EXAMPLES

Example 1

Tumor-specific promoter driven therapy for neuroendocrine tumors: Insulinomas are a rare type of neuroendocrine (NE) tumor, but they are the most common type of tumors arising from pancreatic islet cells. The majority of beta cell tumors are benign, but malignant insulinomas can occur and are highly aggressive tumors that have a 63% 5-year recurrence rate and an average survival of less than 4 years (Proye CA. (1998) Aust. N. Z. J. Surg. 68: 90-100). Moreover, the uncontrolled insulin secretion from these tumors often leads to complications arising from hyperinsulinemia and hypoglycemia. Effective treatment of aggressive and recurring insulinomas is imperative for better clinical outcomes in these patients.

A tumor-specific promoter (insulinoma-associated-1 , INSM1) has been identified that drives a NE-specific tumor marker that was discovered using a human insulinoma subtractive library (Goto et al., (1992) J. Biol. Chem. 267: 15252-15257). The INSM1- promoter regulates expression of a zinc-finger transcription factor that is highly specific to NE tumors (Lan & Breslin (2009) FASEB J. 23: 2024-2033). Insml knockout models have shown that Insml expression is important for the formation of endocrine pancreas and sympatho-adrenal lineage cells during early embryonic development (Gierl et al., (2006) Genes Dev. 20: 2465-2478; Wildner et al., (2008) Development 135:473-481). When the animal reached adulthood, Insml expression could no longer be detected in all normal adult tissues. However, the INSM1 gene was re-activated in most human NE tumors including neuroblastomas, pheochromacytoma, retinoblastomas, medulloblastomas, pituitary carcinomas, small cell lung carcinomas, carcinoid tumors, and insulinomas (Wang et al., (2009) Human Gene Ther. 20: 1308-1318).

The original I NSM 1 -promoter loses tumor specificity when used in an adenoviral vector delivery system. Through a series of modifications using the HS4 chicken β-globin insulator and neuron-restrictive silencer elements (NRSEs) to insulate the promoter against viral enhancers, the modified INSM1 promoter was able to retain specificity to drive adenoviral expression specifically in NE tumors (Akerstrom et al., (2012) Cancer Gene Ther. 19: 828-838).

To take advantage of the NE tumor specificity of the INSM1 promoter, an oncolytic adenoviral vector has been constructed that can express both herpes simplex virus thymidine kinase (HSV-tk) and the Δ24Ε1Α gene with an internal ribosomal entry site (IRES) to act as a therapeutic vector in combination with ganciclovir (GCV) treatment (Moolten et al., (1990) Human Gene Ther 1 : 125-134).

The Δ24Ε1Α mutant is a mutated form of the adenovirus E1A gene essential for viral replication. Due to a 24-base pair deletion, the E1A protein loses its ability to bind retinoblastoma (Rb) protein, resulting in a virus that relies on Δ24Ε1Α for replication solely in Rb negative cancer cells (Whyte et al., (1988) Nature 334: 124-129). The specificity of the Δ24Ε1Α mutant along with the specificity of the INSM1 promoter creates a dual layer of safety for the adenoviral vector. The Ad-INSM1 p- HSV-tk (K5), Ad-I NSM 1 p-A24E1A-IRES- HSV-tk, constructs and the combination of these two vectors were tested to treat insulin secreting tumors using both in vitro cell culture and in vivo xenograft mouse models.

Cloning the INSM1 -promoter driven adenoviral constructs. The Ad-I NSM 1 p-HSV-tk construct (K5) contained a modified INSM1 promoter with a full HS4 chicken β-globin insulator sequence (1.2 kb) upstream and two tandem NRSE sequences downstream (Fig. 1 E) (Akerstrom et al., (2012) Cancer Gene Ther 19:828-838). To generate a new bicistronic oncolytic adenoviral vector, extra space was required for the addition of the IRES element and the Δ24Ε1Α gene. The full HS4 chicken β-globin insulator was replaced with 2 copies of the core insulator sequence (250 bp) (SEQ ID NO: 2) to shorten the modified INSM1 promoter by approximately 700 base pairs (Fig. 1 D). This new second generation modified INSM 1 promoter was able to retain the same tumor specificity as the first generation modified INSM1 promoter.

The conditionally replicating Ad-INSM1p-AE1A-IRES-HSV-tk vector can replicate in an I NSM1 -positive cell after infection. To test the ability of the conditionally replicating virus to replicate inside an I NSM 1 -positive cell, a viral titer assay was performed by infecting RIN cells with Ad-INSM1 p-AE1A-IRES-HSV-tk, the non-replicating Ad-INSM1 p-HSV-tk (K5), or a combination of the two viruses (Fig. 12A). As negative control, virus was added to a blank well without cells. After 3 days, the viral titer for the conditionally replicating virus or the viral combination increased by approximately 3-4-fold as compared to the negative or K5 control. The non-replicating virus displayed no significant change in viral titer in a cell-free medium. The comparison of HSV-tk expression in non-oncolytic versus oncolytic adenoviral vectors. To determine the viral protein expression levels of the replicating Ad-I NSM 1 p-AE1A-IRES- HSV-tk virus and the non-replicating Ad-I NSM 1 p-HSV-tk virus (K5), Western blot analysis was conducted to detect expression of HSV-tk and Δ24Ε1Α after infection in RIN, Ml N, βΤΟ- 1 (INSM1-positive cells), and Panc-1 cells (I NSM 1 -negative cells). Results showed that Ad- INSM1 p-AE1A-IRES-HSV-tk expressed lower levels of HSV-tk than the non-replicating Ad- INSM 1-HSV-tk virus (Fig 12B, 12C). As expected HSV-tk and Δ24Ε1Α protein expression are not detected in Panc-1 cells. The difference in HSV-tk expression between the two viruses was hypothesized to be due to the location of the HSV-tk gene downstream of the IRES element sequence in the replicating virus. Therefore, to improve the HSV-tk expression levels while retaining replication competency, the non-replicating Ad-INSM1 p-HSV-tk virus and the conditionally replicating Ad-INSM1 p-AE1A-IRES-HSV-tk virus were mixed at a ratio of 1 : 1.

Western blots showed that HSV-tk expression was higher in the group treated with the virus combination than the Ad-INSM1 p-HSV-tk virus alone (Fig. 12B, 12D), even though the total number of infectious particles remained constant at the start of the infection in both groups. Comparison of Δ24Ε1Α expression in the combination virus versus the Ad-INSM1 p- ΔΕΙΑ-IRES-HSV-tk virus alone revealed that expression of Δ24Ε1Α was reversed in intensity between RIN and βΤΟ-1 cells (Fig. 12B, 12D). Since adenovirus type 5 infection depends primarily upon expression of the coxsackievirus and adenovirus receptor (CAR) on the cell surface, the low thymidine kinase expression in MIN cells may be due to the low expression of CAR, limiting viral entry into the cells (Fig. 12E).

Oncolytic virus in combination with Ad-INSM1p-HSV-tk increased efficacy of insulinoma killing. Since it was established that a combination of the Ad-INSM1 p-HSV-tk and Ad- INSM1 p-AE1A-IRES-HSV-tk viruses can increase suicide gene expression and viral titer, the efficacy of this combination viral therapy in endocrine and non-endocrine pancreatic cells was examined. A cell survival assay was performed using two different treatment plans to separate the viral oncolytic effect and the HSV-tk/GCV killing.

In the first treatment plan, virus titer was increased (MOI: 0-100) while the GCV concentration was kept constant (100 μΜ). As viral concentration increased, a dose- dependent increase in cell killing was observed in I NSM1 -positive insulinoma cells infected with Ad-INSM1 p-HSV-tk, Ad-INSM1 p-AE1A-IRES-HSV-tk, and the virus combination (Figs. 13C, 13B, 13D). The original Ad-INSM1 p-HSV-tk virus (K5) alone reached maximum killing at 100 MOI, Ad-INSM1 p-AE1A-IRES-HSV-tk virus alone reached maximum killing at 50 MOI, whereas combination virus reached maximum killing at 10 MOI.

When a non-therapeutic non-oncolytic virus such as Ad-INSM1 p-Luc2 was used, no significant changes in cell survival were observed (Fig. 13A). In all cases, the INSM- promoter driven virus displayed specificity to INSM 1-positive cells, sparing the INSM1- negative Panc-1 cells.

The potency of tumor cell killing across each viral treatment was compared and it was found that the combination of viral therapy reached maximum killing effect at a lower concentration (MOI: 10) than the other two viruses alone (Fig. 13D). The virus combination was able to reach the same level of tumor cell killing with an MOI that was 5-10-fold lower than the conditionally replicating Ad-INSM1 p-AE1A-IRES-HSV-tk or K5 virus alone. This result is consistent with Fig. 12C that the HSV-tk and E1A expression levels were greatly increased in the combination virus group to enhance the killing effects.

To further examine the effects of the therapeutic benefits of the viral vectors of the present disclosure, a second treatment plan was devised to explore the effects of GCV drug action with its activating enzyme HSV-tk. The GCV effect (200 μΜ) was tested without virus or increasing the GCV concentration (0-200 μΜ) at a constant viral MOI of 50. Under this regimen, a significant increase of killing in all virus groups was detected except with the Ad- INSM 1 p-Luc2 control (Fig. 14A). Viral specificity was preserved by the INSM1 promoter, killing only I NSM1 -positive cells and leaving I NSM1 -negative Panc-1 cells unaffected.

When tumor killing from either virus alone or the combination of viruses was compared it was observed that the non-replicating Ad-INSM1 p-HSV-tk virus showed similar efficacy to the conditionally replicating Ad-INSM1 p-AE1A-IRES-HSV-tk virus (Figs. 14C and 14B). When the two viruses were combined, efficacy increased by approximately 2-fold (Fig. 14D). Interestingly, GCV by itself in the inactive form can also be toxic to cells,

demonstrated in particular by the decrease in survival of βΤΟ-1 cells when treated with 200 μΜ GCV in the absence of adenovirus.

The Ad-INSM1p-HSV-tk and Ad-INSM1p-AE1A-IRES-HSV-tk virus combination delayed hyperinsulinemia in vivo. The efficacy of the virus combination in vitro was tested for its function in an in vivo xenograft tumor model. RIN cells were infected with 10 MOI Ad- INSM 1 p-Luc2 for 24 hours in vitro to label the cells for subsequent visualization. RIN tumor cells were injected into 6-8 week old Nu/Nu mice and allowed to establish for a period of 48 hours. After this period, Ad-INSM1 p-Luc2 (1 * 10 9 ifu) or a combination of Ad-INSM1 p- ΔΕΙΑ-HSV-tk and Ad-INSM 1 p- HSV-tk (5* 10 8 ifu each) were introduced by direct injection into the same location as injected tumor cells (previously marked with Luc2). Daily GCV treatment was started 3 days after the introduction of the virus.

In the mice treated with the non-therapeutic Ad-INSM1 p-Luc2 virus, blood glucose levels dropped sharply as early as 4 days and continued to drop below 20 mg/dL without recovery (Fig. 15A). Survival of these mice did not last for longer than two weeks, and the luciferase imaging showed the presence of the infected tumor cells for the duration of the experiment (Fig. 15B). In the mice treated with the combination virus, hypoglycemia was delayed in some cases and improved survival to up to 16 days (Fig. 15A). In one case, hypoglycemia was pervented for the duration of the experiment. Using luciferase activity, it was observed that tumor size decreased in mice treated with the combination virus, leading to the prevention or delay of hypoglycemia (Fig. 15C).

Pre-infection with Ad-INSM 1p-HS V-tk and Ad-INSM 1ρ-ΔΕ 1A-IRES-HS V-tk virus

combination displayed cancer specific killing in vivo. To test the viral vector with an alternative delivery method, RIN cells were pre-infected with 10 MOI of the therapeutic combination viruses of the disclosure before injecting them subcutaneously into Nu/Nu mice (Fig. 16). In the control group treated with the non-therapeutic Ad-INSM1 p-Luc2 virus, blood glucose levels began to decrease by day 4 post-injection and continued to decline rapidly without recovery. In contrast, mice in the group treated with either the Ad-INSM 1 p-AE1A- IRES-HSV-tk virus alone or the combination virus and GCV (from day 3) did not experience significant changes in blood glucose for the duration of the experiment, suggesting that HSV- tk/GCV suppresses insulinoma tumor growth and prevents hypoglycemia.

Discussion

Originally, the study focused primarily on the efficacy of the newly constructed conditionally replicating Ad-INSM1 p-AE1A-IRES-HSV-tk virus in comparison with the previously tested non-replicating Ad-INSM 1 p-HSV-tk vector. However, Western blot analysis revealed that while the Ad-INSM1 p-AE1A-IRES-HSV-tk construct expressed high amounts of Δ24Ε1Α protein, it expressed significantly less HSV-tk protein than the Ad- INSM 1 p-HSV-tk construct (K5).

While not wishing to be bound by anyone theory, this decrease in HSV-tk expression can be due to its dependency on the IRES element sequence. Other studies have shown that an IRES-dependent second open reading frame can have significantly lower expression than that of the first gene in a bi-cistronic expression cassette (Mizuguchi et al., (2000) Mol. Ther. 1 : 376-382). To circumvent this inefficiency, a combination of both the Ad-INSM1 p- HSV-tk and Ad-INSM1 p-AE1A-IRES-HSV-tk vectors was used. The co-infection of both viruses into the same cells would not only enhance the non-replicating Ad-INSM1 p- HSV-tk expression, but also retain the replicating capacity due to the presence of Δ24Ε1 A. Using cell survival assays, it was demonstrated that the combination virus displayed a higher efficacy in tumor cell killing as compared to either viral vector alone.

Both viral vector-infected cell lysis and GCV treatment decreased cancer cell survival in a dose-dependent manner. Under conditions of constant viral concentration, the HSV-tk expressing viruses showed higher tumor cell killing at higher GCV concentrations, while non- therapeutic Iuciferase2 virus without HSV-tk showed no effects on cell survival. Conversely, under conditions where GCV concentration is constant and viral concentration is increased, replicating viruses displayed more efficient killing than non-replicating virus as virus MOI increased. When combined, the two groups of viruses, there was a synergistic effect that could result from the increase of viral titers and the HSV-tk suicide gene expression.

In the subcutaneous xenograft tumor model, a direct intra-tumoral injection delivery method it was determined whether the therapeutic virus combination would have any effect on tumor growth. When the tumor-bearing mice were followed, it was found that the resulting hyperinsulinemia from the engrafted tumors had a profound effect on the physiology of tumor bearing animals. As early as 7 days post-tumor cell injection, blood glucose levels dropped below 20 mg/dL and mice treated with non-therapeutic virus (control) did not survive longer than 2 weeks. In the mice treated with the combination virus, hyperinsulinemia was prevented in one subject and was delayed in two subjects by up to 16 days.

The sharp and drastic hyperinsulinemia effect caused difficulties in the timing of the treatment. The tumors did not grow large enough to be visible for injection even as blood glucose dropped significantly during the first few days after the tumor cell injection.

Therefore, it was necessary to inject the viruses early at the same location that the tumor cells had been injected. Most likely, the therapeutic effect was hindered by a less effective delivery of viruses to the tumor cells.

To overcome the abrupt decrease in blood glucose and to treat the tumors in sufficient time, pre-infection of the tumor cells prior to injection was used. This method of treatment provides evidence that the INSM1 promoter is an excellent candidate for driving gene expression to treat insulinomas.

Accordingly, INSM 1 promoter-driven adenoviral vectors like the Ad-INSM1 p-HSV-tk and Ad-INSM1 p-AE1A-IRES-HSV-tk viral combination can be advantageous reducing the proliferation or effects of insulinomas. In cases where patients have recurring insulinoma after surgical resection, an alternative method of treatment is required.

Materials and methods

Cell lines and reagents. The insulinoma cell line βΤΟ-1 was cultured in a Dulbecco Modified Eagle Medium (DMEM, low glucose) supplemented with 10% fetal bovine serum, 50 μg/mL penicillin, and 0.25 μg/mL streptomycin at 37 °C under 5% carbon dioxide. MIN, RIN, and Panc-1 cells were obtained from American type culture collection and cultured in the same medium except that the Panc-1 cells were cultured in high glucose DMEM.

Generation of oncolytic viruses. The pGL3-INSM1 p plasmid contains the modified INSM1 promoter and was digested and ligated with the Δ24Ε1Α and HSV-tk genes linked by an internal ribosomal entry site (IRES). To clone the Δ24Ε1Α gene, site directed mutagenesis was performed on an existing E1A gene in the pJet plasmid to delete 24 base pairs from the original sequence. The HSV-tk gene was obtained from the pWS plasmid while the IRES element sequence was subcloned from the pi RES element vector (Clontech, cat. no.

631605). After ligation, the INSM1 p-A24E1A-IRES-HSV-tk fragment was cut and placed into the pShuttle plasmid, linearized, and electroporated into BJ5183-AD-1 cells for

recombination. The recombinant adenoviral DNA was transfected into AD293 cells to produce adenoviral vector and subjected to further amplification. The final viral stock was titered using the AdenoXTiter Assay (Clontech, cat. no. 632250) and purified using CsCI density gradient centrifugation. Western blots analysis. Cells were infected at 10 MOI for 48 h with either Ad-INSM1 p- ΔΕΙΑ-HSV-tk, Ad-INSM1 p-HSV-tk, or a combination of the two viruses at 5 MOI each. Western blot analysis was performed using a rabbit a-HSV-tk antibody (FL-234, Santa Cruz Biotech., cat. no. sc-134475) at a concentration of 1 : 1000 with a secondary HRP-conjugated goat anti-rabbit antibody at a concentration of 1 :10,000. Blots were then imaged with ECL substrate at an exposure time of 1 minute. After stripping of the membrane, mouse a-E1A antibody (BD Pharmingen, cat. no. 554155) was used at a concentration of 1 : 1000 with a secondary HRP-conjugated goat anti-mouse antibody at 1 :10,000. The membrane was then imaged with ECL substrate at an exposure time less than 30 seconds.

Viral titer assay. RIN cells were seeded on a 6-well plate at a density of 2* 10 6 cells/well and infected with Ad-INSM1 p-AE1 A-HSV-tk, Ad-INSM1 p-HSV-tk, or a combination of the two viruses each at a half concentration (1 * 10 6 ). Viral concentrations of either 10 MOI or 200 MOI were used for each virus group. After incubation for 3 days, cells and media were collected and viral particles were released into the media by 4 cycles of freeze-thaw lysis in a methanol dry ice bath. The viruses infected into AD293 cells were titered by

immunostaining using anti-hexon antibody (Abeam, cat. no. Ab8251). The viral titer was determined using the total number of virus particles based on the volume of media that was collected.

Cell survival assay. Cell survival assays using MTS colorimetric dye were conducted using cell lines that were either I NSM1 -positive or -negative: RIN (INSM1 + rat insulinoma), MIN, β- TC1 (both INSM1 + mouse insulinoma), and Panc-1 (INSM1 " pancreatic epithelioid carcinoma). These cells were plated onto a 96-well plate at a density of 10 5 cells per well. To assess the efficacy of the combination virus versus either the non-replicating virus or the replicating virus alone, a cell survival assay was conducted using the following virus treatment groups: Ad-INSM1 p-Luc2, Ad-INSM1 p-HSV-tk, Ad-INSM1 p-AE1A-IRES-HSV-tk and a combination of Ad-INSM1 p-HSV-tk and Ad-INSM1 p-AE1A-IRES-HSV-tk. These viruses infected cancer cells at a range of 0-200 MOI. After an incubation of 3 days, GCV was added to the cells at a constant 100 μΜ concentration across all viral groups and MOI ranges for five additional days. Afterwards, cell survival was assessed using CELLTITER 96.RTM AQueous One Solution Cell Proliferation Assay (Promega, cat. no. G3582) and read at 490 nm on a Bio-Tek Synergy-HT Microplate Reader. Results were plotted as relative absorbance (relative to control sample with no virus) against increasing viral MOI. Samples were conducted in six wells each and statistical significance was determined using the students T-test with a threshold of p < 0.05.

For a second experiment, the same viral constructs were used to infect cancer cells at a constant 50 MOI. After incubation with the virus for 3 days, GCV was added to the cells at concentrations ranging from 0-200 μΜ. The combination of virus and GCV prodrug was then incubated for five additional days. As control, samples with no virus and either no GCV or 200 μΜ GCV were used to assess the effects of GCV toxicity on cell survival. After the treatment period, cell survival was assessed using CELLTITER 96.RTM AQueous One Solution Cell Proliferation Assay (Promega, cat. no. G3582) and read at 490 nm on a Bio- Tek Synergy-HT Microplate Reader. Results were plotted as relative absorbance (relative to the control sample with no virus and no GCV) against increasing GCV concentration.

Samples were conducted in six wells each and statistical significance was determined using the students T-test with a threshold of p < 0.05.

Mouse xenograft tumors. RIN cells were infected with 10 MOI Ad-INSM 1 p-Luc2 for 24 hours in vitro. After infection, a total of 5x 10 7 cells in a 1 :5 dilution of Matrigel (Corning, cat. no. 354248) in PBS were injected subcutaneously into the right flank of 8 to 10 week old Nu/Nu mice. After a period of 48 hours, Ad-INSM1 p-Luc2 (1 *10 9 ifu) or a combination of Ad- INSM1 p-AE1A-HSV-tk and Ad-INSM1 p-HSV-tk (5 χ 10 8 ifu each) were introduced in the same location as tumor cells where were injected. After 3 days post-injection, daily ganciclovir treatment was administered via intraperitoneal injection. Blood glucose was monitored daily.

To perform the imaging analysis for luciferase activity, D-luciferin substrate (Biosynth, cat. no. L-8220) was prepared at a concentration of 15 mg/ml and injected intraperitoneal ly into mice at a dose of 150 mg/kg. Once injection was completed, the mice were

anesthetized in an isofluorane chamber (2-4% by inhalation) before being transferred to a Kodak In-Vivo Multispectral FX imager (Carestream Health, Rochester, NY). Using the imager's software, luminescence was acquired with a 10 minute exposure with the mice in a supine position and an X-ray image of the mice in the same position was acquired with a 30 second exposure. Imaging was performed every 3 days. To generate the complete image, the luminescence acquisition was converted into a rainbow intensity scale and

superimposed onto the X-ray acquisition using ImageJ software (National Institutes of Health).

In a second experiment, RIN cells were first infected with Ad-INSM1 p-Luc2, Ad- INSM1 p-AE1A-HSV-tk, or a combination of Ad-INSM1 p-AE1A-HSV-tk and Ad-INSM1 p-HSV- tk at 10 MOI for 24 hours. After infection, 10 8 of the pre-infected cells were suspended in a 1 :5 dilution of Matrigel (Corning) in PBS and injected subcutaneously into the right flank of 8 to 10 week old Nu/Nu mice and monitored for blood glucose levels in a similar fashion. After 3 days post-injection, daily ganciclovir treatment was administered via intraperitoneal injection and blood glucose levels were monitored daily.

Statistical Analysis. Values were corrected and expressed relative to a control group. All experiments were repeated three times. Results are presented as mean ± SEM. Statistical analysis was performed using wither the Student's t-test when only two groups were in the experiment or by an one-way ANOVA comparison of multiple groups using the Tukey- Kramer test with differences at p value of less than 0.05 being considered significant.

Example 2

Detection of neuroendocrine tumors using promoter-specific secreted Gaussia luciferase: Neuroendocrine (NE) carcinomas are rare neoplasms that can develop into highly malignant and life-threatening tumors (Rindi & Wiedenmann (2012) Nat. Rev. Endocrinol. 8: 54-64; Jann et al., (201 1) Cancer 117: 3332-3341). While they share a number of genetic and phenotypic traits, NE carcinomas comprise a very heterogeneous population of tumor types that can arise in various organs throughout the body. The most common of these cancers include neuroblastomas, retinoblastomas, medulloblastomas, pituitary carcinomas, small cell lung carcinomas, and carcinoid tumors, encompassing a broad spectrum of tumors that have so far required multiple detection and treatment methods (Brodeur et al., (1993) J. Clin. Oncol. 1 1 : 1466-1477; Hayes et al., (1983) J. Pediatr. 102: 59-62; Oyharcabal-Bourden et al., (2005) J. Clin. Oncol. 23: 4726-4734; Packer et al., (1999) J. Clin. Oncol. 17: 2127-2136; Richardson et al., (1993) Semin. Oncol. 20: 105-127). Despite their differences, many of these tumors express common tumor-specific markers that can identify them as NE cancers (Mountain, C. F. (1978) Sem/A7. Oncol. 5: 272-279; Argiris & Murren, (2001) Cancer J. 7: 437-447). Consequently, early detection of these tumor markers can lead to better treatment response and outcomes.

The INSM1 gene encodes a NE tumor-specific marker that was discovered using an insulinoma subtractive hybridization screen (Goto et al., (1992) J. Biol. Chem. 267: 15252- 15257; Lan et al., (1993) Cancer Res. 53: 4169-4171). The I NSM1 -promoter regulates the expression of INSM 1 , a transcription factor with a zinc-finger DNA binding domain that is highly specific for NE tumors (Breslin et al., (2002) Nucleic Acids Res. 30: 1038-1045).

Through an Insml knockout mouse model, Insml transcription factor was found to be important in the formation of endocrine pancreas and sympatho-adrenal lineage during development (Gierl et al., (2006) Genes Dev. 20: 2465-2478; Wildner et al., (2008)

Development 135: 473-481). Most interestingly, INSM1 expression was discovered to be restricted to the embryonic peripheral and central nervous system, specifically in the cells of neuroendocrine origin (Farkas et al., (2008) Neuron 60: 40-55). The expression pattern was detected in the embryonic tissues of pituitary, pancreas, stomach, duodenum, thymus, adrenal glands, brain, and spinal cord, which were all found to be Insml -positive at E15.5 in mice (Xie et al., (2002) Genomics 80: 54-61 ; Mellitzer et al., (2006) EMBO J. 25: 1344- 1352). However, INSM1 is silenced in normal adult tissues, but reactivated in most of the human NE tumors, including neuroblastoma, medulloblastoma, pheochromocytoma, small cell lung carcinoma, insulinomas, pituitary tumors, carcinoid tumors, medullary thyroid carcinoma, and retinoblastoma (Lan & Breslin (2009) FASEB J. 23: 2024-2033). Therefore, INSM1 is a NE-specific tumor marker.

To assist with the detection of NE tumors despite their heterogeneous population, the I NSM1 -promoter's specificity in NE tumors was used to drive the expression of a

downstream Gaussia luciferase gene. Secreted luciferases like Metridia or Gaussia luciferase have been shown to be highly luminescent, exhibiting 2-4-fold higher signal than Renilla or firefly luciferases (El-Amouri et al., (2013) Mol. Biotechnol. 53: 63-73;

Koutsoudakis et al., (2012) PLoS One 7: e53254). INSM1 p-/Wef and INSM1 p-Gau reporter vectors to measure the INSM1 promoter activity in NE tumors were constructed. In vitro cell lines and xenograft human tumor cultured cells revealed positive luciferase secreted from NE tumors. In addition, combining the INSM1 p-A24E7 \ and INSM1 p-Gai luciferase vectors increased the sensitivity of secreted Gaussia in vivo. The A24E1A gene, a mutant form of the adenovirus E1A gene with a 24 base pair deletion, is inactive in retinoblastoma (Rb) protein expressing cells and active in Rb negative cancer cells (Whyte et al., (1988) Nature 334: 124-129). The cancer specificity from the modified INSM1 promoter and the A24E1A gene create a dual layer of safety against non-specific expression.

Materials and methods

Construction of adenoviral vectors. The Ad-INSM 1 p-/Wef construct was cloned using an original pGL3-INSM1 p vector that contained the modified I NSM1 -promoter with HS4 insulator upstream and 2X NRSE downstream (Akerstrom et al., (2012) Cancer Gene Ther. 19: 828-838). The Metridia luciferase gene was excised from the pMet-Reporter vector (Clontech, Mountain View CA) and ligated downstream of the modified I NSM 1 -promoter in pGL3. The pGL3 vector was cut to release the I NSM1 p-/Wef fragment, which was then ligated into the pShuttle plasmid (Agilent Technologies, Santa Clara CA) for adenoviral vector.

The Ad-INSM 1 p-Gai/ and Ad-INSM1 p-A24E7A constructs were cloned using the modified I NSM1 -promoter on the pGL3-INSM1 p vector, created by shortening the full insulator sequence into two copies of the core HS4 insulator. The Gaussia luciferase gene was obtained from the pMCS-Gai/ss/ ' a-Dura Luc vector (ThermoFisher Scientific, Waltham MA) and ligated downstream of the I NSM 1 -promoter to create pGL3-INSM1 p- Gau. To clone the Δ24Ε1Α gene, site directed mutagenesis was performed on an existing E1A gene in the pJet plasmid (ThermoFisher Scientific, Waltham MA) to delete 24 base pairs from the original sequence. This Δ24Ε1Α gene was then cloned into the pGL3 vector to form pGL3- INSM1 p-424EfA Both the INSM1 p-Gau and the \l·^SM^ -Δ24E1A fragments were excised from their vectors and placed into the pShuttle plasmid. The Ad-SV40-Luc2 construct was generated by excising the SV40 promoter from the pSEAP2-Control vector (Clontech) and ligated upstream of the Luc2 reporter gene in the pGL4.10 vector (Promega, Madison Wl). The SV40- .L C2 fragment was cloned into the pShuttle vector. The pShuttle plasmid was linearized and electroporated into BJ5183-AD-1 cells (Agilent Technologies, Santa Clara CA) to undergo recombination. After selection for the recombinants, linear adenoviral DNA was transfected into AD293 cells (Agilent Technologies, Santa Clara CA) using FuGENE 6 reagent (Promega, Madison Wl). The virus was amplified onto forty 150 mm tissue culture dishes and purified by CsCI gradient. This purified virus was then titered using the Adeno-X- Rapid Titer Kit (Clontech, Mountain View) and stored at -80°C. All sequences in the cloning process were verified through DNA sequencing.

In vivo luciferase imaging. Nu/Nu mice aged 8-10 weeks old received intravenous tail vein injection of either the modified first generation Ad-I NSM 1 p-Luc2, the second generation Ad- INSM1 p- -L c2, or unmodified Ad-I NSM 1 p-/.i c2. The viruses were prepared in phosphate- buffered saline at a concentration of 10 10 ifu per ml and 100 μΙ of the viral solution was delivered slowly into the tail vein via a 27 gauge needle. To perform the imaging analysis for luciferase activity, D-luciferin substrate (Biosynth, Itasca IL) was prepared at a concentration of 15 mg/ml and injected intraperitoneal^ into mice at a dose of 150 mg/kg. Once injection was completed, the mice were anesthetized in an isofluorane chamber (2-4% by inhalation) before being transferred to a Kodak In-Vivo Multispectral FX imager (Carestream Health, Rochester NY). Using the imager's software, luminescence was acquired with a 10 minute exposure and an X-ray image of the mice in the same position was acquired with a 2 minute exposure. Imaging was performed 48 hours after virus injection and periodically up to 28 days. To generate the complete image, the luminescence acquisition was converted into a rainbow intensity scale and superimposed onto the X-ray acquisition using ImageJ software (National Institutes of Health, Bethesda MD). For the NE tumor imaging, H1 155 NE lung tumor cells (1 x 10 7 ) were pre-infected with the second generation modified Ad-INSM1 p- Luc2 virus (50 MOI) for 24 hours and injected subcutaneously into the right hind flank of nude mice (n=3). After one week, the tumor growth was evidenced and imaged to show the modified INSM1 promoter specificity.

In vitro Metridia and Gaussia luciferase secretion assay. Cells were seeded in a 96-well plate at a density of 10,000 cells per well. After incubation at 37°C and 5% C0 2 for 1 h, cells were infected with either no virus (negative control), Ad-INSM1 p-/Wef (0 to 50 MOI), Ad-

INSM1 p-Gau (0 to 50 MOI), or Ad-SV40-/_i/c2 (5 MOI). Infected cells were then incubated at 37°C, 5% C0 2 for 24 h. After incubation, each well was washed gently with 1 * PBS and replaced with fresh media for another 24 hours. Fifty microliters of media per well were transferred to a 96-well white microplate. Luminescence was detected using the Pierce Gaussia Luciferase Glow Assay kit (ThermoFisher Scientific, Waltham MA), and read on a TopCount NXT Microplate Scintillation and Luminescence Counter. The adenoviral infection efficiency was determined by normalization (ratio) with intracellular luciferase (Ad-SV40- Luc2) using the Dual-Glo Luciferase Assay System (Promega, Madison Wl) and read on a TopCount NXT Microplate Scintillation and Luminescence Counter. To test the effects of Ad-INSM1 p-Gai/ in combination with the Ad-INSM1 p-A24£7 \ conditionally replicating adenovirus, cells were infected with a combination of 10 MOI Ad-INSM1 p- Gau and Ad- \l·^SM^ - A24E1 A for a total of 20 MOI (2 χ 10 5 ifu). Media was collected 2, 4, and 6 days (20 μΙ each day) after infection to determine secreted luciferase activity. Luminescence was detected using the Pierce Gaussia Luciferase Glow Assay kit (ThermoFisher Scientific, Waltham MA), then read on a TopCount NXT Microplate Scintillation and Luminescence Counter. The Student's T-test with a threshold of p<0.05 was used to determine statistical significance. This process was repeated with the Ad- 1 NSM1 p- Gau infected cells to determine the Ad-INSM1 p-Gai//Ad-SV40-/_i/c2 ratio.

Xenograft human tumor culture assay. Xenograft tumors were prepared by injecting human tumor cells (1 x 10 7 ), such as HeLa, U87, D283, UMC-1 1 , SK-NBe(2), H1 155, and H69 subcutaneously into the right hind flank of nude mice. Tumor tissues were harvested and frozen (-80 °C) in RPMI 1640 culture medium with 10 % DMSO. The cultured tumor cells were prepared by rapidly thawing in a 37°C water bath and subsequent mincing into small sections approximately 1 mm 3 in size. The minced tissues were then centrifuged at 250*g for 1 minute and incubated in 2.5% trypsin for a total of 30 minutes at 37°C and 2 ml growth media was added to neutralize the trypsin. The trypsinized tissues were then filtered through a 70 μηι sieve and centrifuged again at 250*g for 5 minutes. The 96-well clear- bottom plates were coated with 75 μΙ per well of a 1 :6 dilution of Matrigel in RPMI growth media and then incubated for 30 minutes at 37°C. The tumor cells (10,000 cells) were re- suspended in RPMI media and added to each Matrigel coated well. Cells from each tumor were infected with Mefr/d/a-luciferase and Ad-S 0- Luc2 at 37°C for 24 hours. The ratio of Metridia/Luc2 luciferase was calculated and averaged using RFU from 50 μΙ media per well. Detection of serum Gaussia luciferase in vivo. Eight week old Nu/Nu mice (National Cancer Institute, Bethesda MD) were injected with H1 155 NE lung tumor cells (1 χ 10 7 )

subcutaneously into the right hind flank. Tumors were allowed to establish until tumor size grew to at least 0.1 cm 3 in volume. The mice were injected intra-tumorally with 1 * 10 9 ifu of Ad-INSM1 p-/_i/c2 virus, or a combination of 5* 10 8 ifu of Ad- IN SM 1 p- Gau and Ad-INSM 1 p- A24E1A (for a total of 1 *10 9 ifu). To detect the Gaussia expression in the bloodstream, 100 μΙ of blood was drawn at 3, 6, 9, and 12 days after virus injection. All animal experiments were performed in accordance with the approved protocol from the Institutional Animal Care and Use Committee, Louisiana State University Health Sciences Center New Orleans. The collected blood was allowed to clot for 30 minutes at room temperature and centrifuged at 2,000 g for 10 minutes. Serum was collected from the supernatant and diluted with PBS at a 1 : 10 ratio. To detect Gaussia luciferase in the serum, 50 μΙ of the diluted serum from each sample was added to a flat bottom 96 well plate for the Gaussia luciferase assay.

Statistical Analysis. Values were corrected and expressed relative to a control group. All experiments were repeated three times. Results are presented as mean ± SEM. Statistical analysis was performed using wither the Student's t-test when only two groups were in the experiment or by an one-way ANOVA comparison of multiple groups using the Tukey- Kramer test with differences at p value of less than 0.05 being considered significant.

Cloning the INSM1 -promoter driven adenoviral constructs. To generate an adenoviral vector that is useful for the diagnosis of I NSM1 -positive NE tumors, the first generation modified I NSM 1 -promoter was constructed by inserting a full HS4 insulator sequence upstream of the I NSM 1 -promoter (approximately 1.7 kb) along with two NRSE enhancer sequences in tandem repeats downstream and Luc2 gene (Fig. 12A) (Akerstrom et ai, (2012) Cancer Gene Ther. 19: 828-838). The modified I NSM 1 -promoter drives a downstream Metridia luciferase gene, resulting in the construct Ad-HS4ins-INSM1 p-2xNRSE- Metridia (Ad- INSM1 p-/Wef) (Fig. 12B). A second generation of the modified I NSM1 -promoter was constructed to drive the expression of Gaussia luciferase and Δ24Ε1Α. This promoter was created using two copies of the HS4 core insulator in place of the full insulator sequence. The final constructs Ad-2xHS4Core-INSM1 p-2xNRSE-Gat/ss/a (Ad-INSM1 p-Gau) and Ad- 2xHS4Core-INSM1 p-2xNRSE-424E7A (Ad-INSM1 p-A24EfA), have a modified INSM1- promoter that is -700 bp shorter than that of the promoter in Ad-INSM1 p-/Wef (Fig. 12C, 12D). Ad-S 0- Luc2 vector was constructed as a control vector (Fig. 12E).

It was examined whether an adenoviral vector driven by the modified INSM1- promoter would result in non-specific expression in vivo. Tail vein injection was performed using three viral vectors, the un-modified Ad-INSM1 p-/-i c2 (Fig. 17A), the first generation of the modified Ad-INSM1 p-/_i/c2 (Fig. 17B), and the second generation modified Ad-INSMp- Luc2 (Fig. 17C) injected into non-tumor bearing Nu/Nu mice separately. Therefore, the INSM 1 promoter specificity with or without HS4 insulator and NRSE enhancer sequence was examined. After a period of 2-28 days, luciferase activity was determined via in vivo imaging system after intraperitoneal (i.p.) injection of luciferin substrate using a Kodak In-Vivo Multispectral FX imager. The intravenous injected adenovirus usually harbored in the liver (>90%). In the non-tumor bearing mice that were injected with the original un-modified Ad- INSM1 p- -L c2, it was observed that non-specific luciferase expression occurred and was focused primarily in the liver area. In contrast, both the first and second generation modified Ad-INSM1 p- -L c2 did not exhibit non-specific luciferase expression after luciferin

administration. To further demonstrate that the modified Ad-INSM1 p-/-i c2 virus maintains NE tumor specificity, an Ad-INSM1 p-/-i c2 pre-infected H1 155 NE tumor was established in nude mice and showed tumor imaging by luciferase readily (Fig. 17D). These results determined that the modified I NSM1 -promoter (both first and second generation) is essential in blocking the effects of adenoviral regulatory elements to retain tumor specificity in vivo. Ad-INSM1p-Met displays INSM1 specificity in vitro. An adenoviral vector to express secreted Metridia luciferase specifically driven by the modified I NSM1 -promoter for the detection of NE tumors was constructed. Secreted Metridia was measured in vitro by co- infecting tumor cells with Ad-I NSM 1 p-Met vector (Ad-HS4ins-INSM1 p-2xNRSE-Mefr/cf/a) and Ad-SV40-/-i/c2. The addition of Ad-SV 40- Luc2 virus was used to normalize the infection efficiency using the ratio between extracellular and intracellular luciferase activity.

Both I NSM 1 -negative and I NSM 1 -positive tumor cell lines including lung carcinoma, neuroblastoma, medulloblastoma, pheochromocytoma, and insulinoma were infected with Ad-INSM1 p-/Wef/Ad-SV40-/_i c2 for 48 hours (Figs. 18A-18C). The secreted luciferase activity in the media was readily detected in all of the I NSM 1 -positive cell lines (solid lines). In particular, the I NSM 1 -positive cell lines H82, βΤΟ-1 , and WERI-Rb-1 exhibited the highest Ad-I NSM 1-Met/Ad-SV40-/_i/c2 luminescence ratios. At the highest MOI (50:1), secreted luciferase activity reached higher than two-fold that of the intracellular luciferase activity (Figs 18A-18C). In contrast, the I NSM 1 -negative tumor cell lines (dot lines) showed no secreted Metridia luciferase relative to intracellular luciferase.

To assess the efficacy of I NSM 1 -promoter driven Metridia luciferase adenoviral vector in xenograft human tumors, human tumor cultured cells derived from previously established xenograft tumor were collected and grown in culture. These ex vivo tumor cells were co-infected with Ad-I NSM 1 p-/Wef and Ad-SV40- Luc2 to determine the ratio between extracellular and intracellular luciferase activity. After incubation for 3 days, it was determined that INSM 1-positive cells (UMC-11 , SK-N-Be(2), H1155, H69, except D283) infected by Ad-INSM1 p-/Wef expressed extracellular Metridia luciferase that produced signals as high as 1.6 times the intracellular firefly luciferase (Fig. 19). In contrast, INSM1-negative cells (HeLa and U87) produced low levels of extracellular Metridia luciferase that did not exceed 0.11 times the activity of intracellular firefly luciferase.

INSM1 promoter-driven Gaussia luciferase retains specificity: In preparation for further in vivo assays, a Gaussia luciferase expression vector was preferred due to the increased stability of Gaussia luciferase in vivo. As recent studies have shown, Gaussia luciferase signals were detectable after tail-vein injection in mice, due to its increased temperature stability compared to Metridia luciferase. To determine the specificity of the newly constructed Gaussia luciferase construct Ad-I NSM ρ-Gau, an in vitro luciferase assay was conducted to evaluate whether the Ad-I NSM 1 p-Gai/ vector (Ad-2xHS4Core-INSM 1 p- 2xNRS E-Gaussia) could specifically express Gaussia luciferase in I NSM 1 -positive cell lines in a similar fashion as did the Ad-I NSMp-Mef vector. After co-infection with Ad-I NSM 1 p- Gai//Ad-SV40-/-i/c2, it was possible to see significant secreted luminescent activity in the media of all I NSM1 -positive cell lines (solid lines) as compared to I NSM1 -negative control cell lines (dot lines) (Figs. 20A-20C). In particular, the I NSM1 -positive cell lines βΤΟ-1 , RIN, H82, and H1155 exhibited the highest Ad-INSM1-Gau/Ad-SV40-/_i/c2 ratios. Secreted Gaussia luciferase was not detected in any I NSM 1 -negative cell lines, indicating INSM1 promoter retains specificity in vitro.

Conditional replicating vector (Ad-INSM1p-A24E1A) enhances Gaussia luciferase secretion and sensitivity over time in an in vivo mouse xenograft tumor model. After establishing the specificity of the Ad-INSM1 p-Gai vector, it was determined whether infecting cells with Ad- \NSMp-A24E1A in combination with Ad-INSM 1 p- Gau could increase the secreted luciferase signal. Accordingly, if an I NSM1 -positive cell is infected concurrently with both viruses, Δ24Ε1Α expression from Ad-INSM1 p-A24E7 \ can be utilized by both viruses to facilitate replication. The replication of Ad-INSM1 p-Gai would lead to viral amplification and increased Gaussia luciferase secretion over time.

Therefore, H1 155 NE lung carcinoma cells or SK-N-Be(2) neuroblastoma cells were co-infected with Ad-INSM 1 p-Gau alone (20 MOI) or in combination with Ad-INSM 1 ρ-Δ24Ε 1A at a concentration of 10 MOI each (Fig. 21). Six days after infection, the combination viruses displayed a 2-fold increase in secreted luminescent activity as compared to infection with Ad-INSM ρ-Gau alone. This result suggested that the combination viruses could indeed amplify inside an INSM-positive cell in vitro.

It was analyzed whether the combination of Ad-INSM 1 p- Gau and Ad-INSM1 p-

A24E1A viruses could secrete detectable amount of Gaussia luciferase into the circulation from a tumor bearing animal for an extended period of time. In this experiment,

subcutaneous H1155 tumors (approximately 0.1 cm 3 ) were first established on the right flank of Nu/Nu mice. These tumor-bearing mice (n = 3) were then injected intra-tumorally with either Ad-INSM1 p-/_i/c2 or the Ad-INSM1 p-Gai/ and Ad-I NSM 1 p-A24E 1A virus combination at a total concentration of 1 * 10 9 ifu (Fig. 22). After infection with the virus combination for 6 days, detectable luciferase signal was observed in the serum (p < 0.01). The signal increased in intensity by day 9 and lasted up to 12 days, the humane endpoint for the tumor bearing animals. The Ad-I NSM 1 p- .t c2 infected tumor released no Gaussia luciferase into the circulation and was used as the control.

Although the original INSM1-promoter possesses NE-tumor specificity, it was found that the promoter loses its specificity when used in an adenoviral setting. It has been demonstrated that an I NSM 1 -promoter driven adenoviral reporter construct displayed nonspecific expression after tail vein injection in an in vivo mouse model (Akerstrom et al., (2012) Cancer Gene Ther. 19: 828-838). It was hypothesized that this loss of specificity was due to the presence of overpowering viral enhancers that were otherwise not present in normal cells. To override these adenoviral regulatory elements, an insulator sequence derived from the HS4 chicken β-globin insulator was placed upstream of the INSM1- promoter to block effects from any viral enhancers. In addition, two copies of the neuron- restrictive silencer element (NRSE), a regulatory element with dual functions to silence the INSM 1 promoter in non-neuronal cells while enhancing it in neuronal cells, were placed downstream of the promoter. Once these elements were added, the modified INSM1- promoter was able to retain its high specificity in an adenoviral vector (Akerstrom et al., (2012) Cancer Gene Ther. 19: 828-838).

To further improve upon this original design, the 1.2 kb full insulator sequence has been replaced with two copies of the HS4 core insulator (250 bp χ 2) to create the second generation modified INSM1 promoter. Although the 1.2 kb full insulator sequence has been well characterized functionally, the 250 bp core insulator was observed to exhibit the same protective activity as the full sequence (Aker et al., (2007) Hum. Gene Ther. 18: 333-343). An advantage of switching from a full insulator sequence to the core sequence is that utilization of two copies of the 250 bp core would free approximately 700 bp of space for the assembly of larger transgenic sequences in the viral vector. Essentially, this more compact form of the modified I NSM1 -promoter displays the same NE tumor specificity with the additional advantage of allowing more flexible cloning strategies.

Retaining the specificity of the I NSM1 -promoter in an adenoviral vector has allowed the construction of a Gaussia luciferase reporter vector that can detect the presence of NE tumor in vivo. When paired with a conditionally replicating oncolytic virus, the virus combination allowed for continuous expression of Gaussia luciferase for the duration of the tumor's progression. These results can have a significant impact on monitoring tumor progression during the treatment of patients. Given that the viruses can selectively replicate in NE tumor cells, Gaussia luciferase expression will persist and intensify as the tumor increases in size. Conversely, if treatment of the tumor is successful, luciferase expression in the patient's blood will decrease as tumor size is reduced. Provided is evidence that the Gaussia vector can be used in combination with a treatment protocol to monitor a patient's treatment outcome. An alternative use for this virus during the treatment of a NE tumor would be to discern whether a tumor is removed completely after surgical resection. By injecting the virus combination into the resection site during the surgical procedure, clinicians would be able to monitor the presence of I NSM 1 -positive NE tumor cells based on a Gaussia luciferase readings from the patient's blood. Continuous monitoring of expression levels will allow for a better prognosis in these patients post-procedure by alerting clinicians to an incomplete resection.

Using the Ad-INSM1 p-Gai/ vector in combination with the Ad-INSM1 p-A24EM was discovered to be more advantageous as compared to using Ad-INSM1 p-Gai/ alone. In NE tumor cells infected by the virus combination, Gaussia luciferase expression was significantly higher than that of the Ad-INSM1 p-Gai virus (20 MOI) alone after 6 days post infection, even though the number of infectious units of Ad- IN SM 1 p- Gau (10 MOI) was lower at the start for the combination. This indicates that the addition of Δ24Ε1Α expression in cells infected by the Gaussia virus allowed for conditional replication of the reporter vector. This replication has the potential to significantly increase the copy number of the virus over several days, leading to an increase in sensitivity of Gaussia luciferase detection. Therefore, the most efficient method of increasing the sensitivity of infection seems to involve utilization of conditionally replicating viruses, as opposed to simply increasing the infectious units during administration of the virus.

Taken together, the Ad-INSM1 p-Gai virus has the potential to be an easy-to-use and highly sensitive tool for the detection of NE tumors in the clinical setting. While a viral construct cannot currently be used as a diagnostic tool for the general population, it can be an alternative approach to track the tumor progression in patients with existing NE cancers. Additionally, it could also be used diagnostically in populations where a NE tumor is suspected. In these cases, the virus combination could act as both a diagnostic tool and as a way to monitor tumor progression.

Example 3

Small cell lung carcinomas are a group of highly malignant neuroendocrine (NE) cancers that have poor prognosis due to high rates of growth and metastasis.

Characteristically a diffuse, non-solid tumor, patients with SCLC most often rely on radiation and chemotherapy for treatment. In the case of unresponsive or relapsing tumors, treatment options become limited and median survival is low. To increase treatment options for SCLC patients, it has been possible to generate a conditionally replicating adenovirus that can specifically replicate and express therapeutic genes in NE tumor cells. The expression of these adenoviruses is regulated upstream by an insulinoma-associated-1 (INSM1) promoter, which is silent in normal adult tissues but active in NE tumors and developing NE tissues. By placing the I NSM1 -promoter in an adenoviral construct, the construct can retain tumor specificity and drive expression of a mutated adenovirus E1A gene (Δ24Ε1Α) or the herpes simplex virus thymidine kinase (HSV-TK) gene.

In vitro cell line and subcutaneous mouse tumor models revealed that the INSM1- promoter driven constructs were able to replicate specifically in I NSM1 -positive cells. INSM1 specific HSV-TK expression in combination with ganciclovir treatment displayed dose- dependent tumor cell death in NE tumor cells, leaving INSM1-negative cells unharmed. When combined, the I NSM1 -promoter driven HSV-TK with Δ24Ε1Α, directed co-infected NE tumors to express higher levels of HSV-TK and more efficient tumor suppression compared to the INSM1 p-HSV-TK virus alone. In an orthotopic mouse lung model, SCLC tumors established in the lungs were treated with INSM1 driven adenoviruses by delivery through the nasal passage to

realistically simulate lung cancer progression and treatment. Monitoring of the orthotopic lung tumors by luciferase signal revealed decreased tumor size after infection for up to 20 days. Taken together, I NSM1 -driven conditionally replicating adenoviruses represent a new tool for the treatment of SCLCs, giving clinicians additional options to combat this deadly disease.

Generation and use of oncolytic viruses (as shown in Figs 23-31): The pGL3 INSMI p plasmid contains the INSM 1 promoter and was digested and ligated with the Δ24Ε1Α and HSV-TK genes linked by an internal ribosomal entry site (IRES). To clone the Δ24Ε1Α gene, site directed mutagenesis was performed on an existing E1A gene in the pJet plasmid to delete 24 base pairs from the original sequence. The HSV-TK gene was cut from the pWS plasmid while IRES element was subcloned from the pIRES element vector

(Clonetech). After ligation, the pGL3 vector was cut and the INSM1 p-A24E1A-IRESHSV-TK fragment was placed into the pShuttle plasmid, then linearized and electroporated into

BJ5183-AD-1 cells for recombination. After recombination, adenoviral DNA was transfected into AD293 cells to be grown and amplified. The final virus will be titered using the

AdenoXTiter Assay (Clonetech) and purified using CsCI density gradient centrifugation. Western blot: After viral infection for 48 hours, total protein extracts were isolated from each infected cell line and anti-E1A and anti-HSVTK antibodies were used to detect protein expression. Anti-GAPDH and anti-CAR antibodies were used as control.

Cell survival assays: Cell lines were infected by viruses at increasing multiplicities of infection (MOI), starting at 0 and increasing in orders of magnitude to 0.1 , 1 , and 10 MOI. All cells will be incubated for 5 days, after which cell survival was measured by MTS Assay (Promega). Absorbance at 490 nm was measured and plotted against MOI. Crystal violet staining was performed at 0.1 , 1 , and 10 MOI after infection for 5 days.

Viral titer assay: H1155 cells were plated on 6-well plates at a density of 2x 10 6 cells/well and infected with a viral concentration of 5 MOI. After incubation for approximately 3 days, cells were collected and viral particles released by freeze thaw lysis. The isolated virus were then titered by immunostaining.

Mouse xenograft tumors: A total of 10 9 cells were injected either subcutaneously into the right flank or orthotopically into the left lung of 4 week old Nu/Nu mice. After

establishment of tumor, viruses were introduced by direct injection in a subcutaneous tumor or through nasal inhalation for a lung tumor.

Through western blot, viral constructs where shown to express ΔΕ1Α and HSVTK.

Additionally, the combination virus expressed higher levels of ΔΕ1Α and HSVTK than either virus alone. INSM 1 promoter driven virus limited its expression specifically to INSM1 positive cell lines in vitro, leaving INSM1 negative cells unaffected.

Cell survival assays revealed the combination of ΔΕ1Α and HSVTK was more effective at killing than ΔΕ1Α alone. The combination virus was shown to amplify after infecting an INSM1 positive cell.

The combination viruses displayed effective suppression of subcutaneous tumors. Depending on the aggressiveness of the specific tumor cell, suppression of tumors lasted up to 28 days.

The combination viruses were effective at suppressing orthotopically established lung tumors after administration through the nasal passage. This suppression was observed for up to 20 days using luciferase imaging.

Example 4

Table 1: Sequences used to generate the constructs of the disclosure: