Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NANOCONJUGATES ABLE TO CROSS THE BLOOD-BRAIN BARRIER
Document Type and Number:
WIPO Patent Application WO/2013/040499
Kind Code:
A1
Abstract:
Polyvalent nanoconjugates address the critical challenges in therapeutic use. The single-entity, targeted therapeutic is able to cross the blood-brain barrier (BBB) and is thus effective in the treatment of central nervous system (CNS) disorders. Further, despite the tremendously high cellular uptake of nanoconjugates, they exhibit no toxicity in the cell types tested thus far. This property is critical for therapeutic agent delivery applications for reducing off-target effects.

More Like This:
Inventors:
MIRKIN CHAD A (US)
KO CAROLINE H (US)
STEGH ALEXANDER (US)
GILJOHANN DAVID A (US)
LUCIANO JANINA (US)
JENSEN SAM (US)
Application Number:
PCT/US2012/055635
Publication Date:
March 21, 2013
Filing Date:
September 14, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV NORTHWESTERN (US)
MIRKIN CHAD A (US)
KO CAROLINE H (US)
STEGH ALEXANDER (US)
GILJOHANN DAVID A (US)
LUCIANO JANINA (US)
JENSEN SAM (US)
International Classes:
A61K47/48
Domestic Patent References:
WO2011028847A12011-03-10
WO2011053940A22011-05-05
WO2011028847A12011-03-10
WO2003008539A22003-01-30
WO1997012896A11997-04-10
WO1998039352A11998-09-11
WO1999014226A21999-03-25
WO1999014226A21999-03-25
WO2008098248A22008-08-14
Foreign References:
US20100233084A12010-09-16
US20100047163A12010-02-25
US20100233270A12010-09-16
US20080274202A12008-11-06
US20080213179A12008-09-04
US20100158931A12010-06-24
US20100209425A12010-08-19
US20100233084A12010-09-16
US7238472B22007-07-03
US20030147966A12003-08-07
US20100055018W2010-11-01
US20060040124W2006-10-13
US5432272A1995-07-11
US3687808A1972-08-29
EP1072679A22001-01-31
US4845205A1989-07-04
US5130302A1992-07-14
US5134066A1992-07-28
US5175273A1992-12-29
US5367066A1994-11-22
US5457187A1995-10-10
US5459255A1995-10-17
US5484908A1996-01-16
US5502177A1996-03-26
US5525711A1996-06-11
US5552540A1996-09-03
US5587469A1996-12-24
US5594121A1997-01-14
US5596091A1997-01-21
US5614617A1997-03-25
US5645985A1997-07-08
US5830653A1998-11-03
US5763588A1998-06-09
US6005096A1999-12-21
US5750692A1998-05-12
US5681941A1997-10-28
US7223833B12007-05-29
US5270163A1993-12-14
US5637459A1997-06-10
US5539082A1996-07-23
US5714331A1998-02-03
US5719262A1998-02-17
US4981957A1991-01-01
US5118800A1992-06-02
US5319080A1994-06-07
US5359044A1994-10-25
US5393878A1995-02-28
US5446137A1995-08-29
US5466786A1995-11-14
US5514785A1996-05-07
US5519134A1996-05-21
US5567811A1996-10-22
US5576427A1996-11-19
US5591722A1997-01-07
US5597909A1997-01-28
US5610300A1997-03-11
US5627053A1997-05-06
US5639873A1997-06-17
US5646265A1997-07-08
US5658873A1997-08-19
US5670633A1997-09-23
US5792747A1998-08-11
US5700920A1997-12-23
US20040219565A12004-11-04
US4469863A1984-09-04
US4476301A1984-10-09
US5023243A1991-06-11
US5177196A1993-01-05
US5188897A1993-02-23
US5264423A1993-11-23
US5276019A1994-01-04
US5278302A1994-01-11
US5286717A1994-02-15
US5321131A1994-06-14
US5399676A1995-03-21
US5405939A1995-04-11
US5453496A1995-09-26
US5455233A1995-10-03
US5466677A1995-11-14
US5476925A1995-12-19
US5519126A1996-05-21
US5536821A1996-07-16
US5541306A1996-07-30
US5550111A1996-08-27
US5563253A1996-10-08
US5571799A1996-11-05
US5587361A1996-12-24
US5194599A1993-03-16
US5565555A1996-10-15
US5527899A1996-06-18
US5721218A1998-02-24
US5672697A1997-09-30
US5625050A1997-04-29
US5489677A1996-02-06
US5602240A1997-02-11
US5034506A1991-07-23
US5166315A1992-11-24
US5185444A1993-02-09
US5214134A1993-05-25
US5216141A1993-06-01
US5235033A1993-08-10
US5264562A1993-11-23
US5264564A1993-11-23
US5405938A1995-04-11
US5434257A1995-07-18
US5470967A1995-11-28
US5541307A1996-07-30
US5561225A1996-10-01
US5596086A1997-01-21
US5610289A1997-03-11
US5608046A1997-03-04
US5618704A1997-04-08
US5623070A1997-04-22
US5663312A1997-09-02
US5633360A1997-05-27
US5677437A1997-10-14
US5792608A1998-08-11
US5646269A1997-07-08
US5677439A1997-10-14
US20100044844W2010-08-09
US20020172953A12002-11-21
US6361944B12002-03-26
US6417340B12002-07-09
US6495324B12002-12-17
US6506564B12003-01-14
US6582921B22003-06-24
US6602669B22003-08-05
US6610491B22003-08-26
US6678548B12004-01-13
US6677122B22004-01-13
US6682895B22004-01-27
US6709825B22004-03-23
US6720147B22004-04-13
US6720411B22004-04-13
US6750016B22004-06-15
US6759199B22004-07-06
US6767702B22004-07-27
US6773884B22004-08-10
US6777186B22004-08-17
US6812334B12004-11-02
US6818753B22004-11-16
US6828432B22004-12-07
US6827979B22004-12-07
US6861221B22005-03-01
US6878814B22005-04-12
US20110111974A12011-05-12
Other References:
See also references of EP 2755692A4
PARDRIDGE, NEUROVIROL., vol. 5, 1999, pages 556 - 569
BICKEL ET AL., ADV. DRUG DELIV. REV., vol. 46, 2001, pages 247 - 279
TSUJI ET AL., ADV. DRUG DELIV. REV., vol. 36, 1999, pages 277 - 290
KUSUHARA ET AL., DRUG DISCOV. TODAY, vol. 6, 2001, pages 150 - 156
DEHOUCK ET AL., J. CELL. BIOL., vol. 138, 1997, pages 877 - 889
FILLEBEEN ET AL., J. BIOL. CHERN., vol. 274, 1999, pages 7011 - 7017
LJUBIMOVA ET AL., CHEM. BIOL. INTERACT, vol. 171, no. 2, pages 195 - 203
BONOIU ET AL., PROC. NATL. ACAD. SCI. USA, vol. 106, no. 4, pages 5546 - 5550
PARDRIDGE, CURR OPIN PHARMACOL, vol. 6, 2006, pages 494 - 500
PARDRIDGE, J. NEUROVIR., vol. 5, 1999, pages 538 - 555
RAPOPORT: "Blood Brain Barrier in Physiology and Medicine", 1976, RAVEN PRESS
JOHANSON: "Neuronal Microenvironment", 1988, THE HUMANA PRESS, article "The choroid plexus-arachnoid membrane-cerebrospinal fluid system", pages: 33 - 104
BALIN ET AL., J COMP NEUROL, vol. 251, 1986, pages 260 - 280
BROADWELL ET AL.: "The Blood-Brain Barrier", 1993, RAVEN PRESS LTD, article "Cell biological perspective for the transcytosis of peptides and proteins through the mammalian blood-brain fluid barriers", pages: 165 - 199
BROADWELL ET AL., PROC NATL. ACAD SCI. USA, vol. 78, 1981, pages 7820 - 7824
HAVASHI, VAC. SCI. TECHNOL., vol. A5, no. 4, 1987, pages 1375 - 84
HAYASHI, PHYSICS TODAY, 1987, pages 44 - 60
MRS BULLETIN, January 1990 (1990-01-01), pages 16 - 47
MARINAKOS ET AL., ADV. MATER., vol. 11, 1999, pages 34 - 37
MARINAKOS ET AL., , CHEM. MATER., vol. 10, 1998, pages 1214 - 19
ENUSTUNTURKEVICH, J. AM. CHEM. SOC., vol. 85, 1963, pages 3317
HURST ET AL., ANALYTICAL CHEMISTRY, vol. 78, no. 24, 2006, pages 8313 - 8318
ZHANG ET AL., J AM CHERN SOC., vol. 132, no. 43, 2010, pages 15151 - 15153
ZHANG ET AL., JAM CHERN SOC., vol. 132, no. 43, 2010, pages 15151 - 15153
SUSAN M. FREIERKARL-HEINZ ALTMANN, NUCLEIC ACIDS RESEARCH, vol. 25, 1997, pages 4429 - 4443
"Antisense Research and Application", September 1993, CRC PRESS
COOK, ANTI-CANCER DRUG DESIGN, vol. 30, 1991, pages 585 - 607
TUERK ET AL., SCIENCE, vol. 249, 1990, pages 505 - 859
NIELSEN, SCIENCE, vol. 254, 1991, pages 1497 - 1500
SANGHVI, Y. S.: "Antisense Research and Applications", 1993, CRC PRESS, pages: 289 - 302
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989
KATZ, J. AM. CHEM. SOC., vol. 74, 1951, pages 2238
YAMANE ET AL., J. AM. CHEM. SOC., vol. 83, 1961, pages 2599
KOSTURKO, BIOCHEMISTRY, vol. 13, 1974, pages 3949
THOMAS, J. AM. CHEM. SOC., vol. 76, 1954, pages 6032
ZHANG ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 74 - 75
ZIMMERMANN ET AL., J. AM. CHEM. SOC., vol. 124, 2002, pages 13684 - 13685
YAN ET AL., RNA BIOL., vol. 6, no. 3, 2009, pages 316 - 320
SONG ET AL., CHEM ING ENGL, vol. 48, no. 48, 2009, pages 9143 - 9147
CRAWFORD ET AL., BRIEFINGS IN FUNCTIONAL GENOMICS AND PROTEOMICS, vol. 2, no. 1, 2003, pages 72 - 79
KOPYLOV ET AL., MOLECULAR BIOLOGY, vol. 34, no. 6, 2000, pages 940 - 954
MOLEKULYAMAYA BIOLOGIYA, vol. 34, no. 6, 2000, pages 1097 - 1113
HARRIS ET AL., NAT REV DRUG DISCOV., vol. 2, 2003, pages 214 - 21
MESMAEKER, CURRENT OPINION IN STRUCTURAL BIOLOGY, vol. 5, 1995, pages 343 - 355
MARTIN ET AL., HELV. CHIM. ACTA, vol. 78, 1995, pages 486 - 504
REMINGTON'S PHARMACEUTICAL SCIENCES, 1980
JONES ET AL., PHARM RES., vol. 24, no. 9, 2007, pages 1759 - 1771
JEFFERIES ET AL., NATURE, vol. 312, 1984, pages 162 - 163
DUFFY ET AL., BRAIN RES, vol. 420, 1987, pages 32 - 38
GOLDEN ET AL., J CLIN INVEST, vol. 99, 1997, pages 14 - 18
GAILLARD ET AL., EXPERT OPIN DRUG DELIV, vol. 2, 2005, pages 299 - 309
GAILLARD ET AL., INT CONGRES SERIES, vol. 1277, 2005, pages 185 - 198
FENG ET AL.: "Drug Delivery to the Central Nervous System", vol. 45, 2010, pages: 15 - 34
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ZHANGMADDEN, GENOME RES., vol. 7, 1997, pages 649 - 656
STEGH ET AL., GENES DEV., vol. 21, 2007, pages 98 - 111
STEGH ET AL., PROC NATL ACAD SCI U.S.A., vol. 105, 2008, pages 10703 - 8
Attorney, Agent or Firm:
ALLIKIAN, Michael J. et al. (Gerstein & Borun LLP233 S. Wacker Drive,6300 Willis Towe, Chicago IL, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A composition comprising a nanoconjugate, the nanoconjugate comprising a polynucleotide that is sufficiently complementary to a target polynucleotide which encodes a polypeptide specifically expressed in a central nervous system (CNS) disorder, the nanoconjugate having the ability to cross the blood-brain barrier (BBB).

2. The composition of claim 1, further comprising a targeting moiety.

3. The composition of claim 1 or claim 2, further comprising a therapeutic agent.

4. The composition of claim 3 wherein the therapeutic agent is temozolamide.

5. The composition of any one of claims 1-4 wherein the disorder is caused by aberrant gene expression.

6. The composition of any one of claims 1-5 wherein the disorder is acute.

7. The composition of claim 6 wherein the acute disorder is selected from the group consisting of focal brain ischemia, global brain ischemia, brain trauma, spinal cord injury, acute infections, status epilepticus, migraine headache, acute psychosis, suicidal depression, acute anxiety/phobia and injury related maladies.

8. The composition of any one of claims 1-5 wherein the disorder is chronic.

9. The composition of claim 8 wherein the chronic disorder is selected from the group consisting of chronic neurodegeneration, retinal degeneration, depression, chronic affective disorders, lysosmal storage disorders, chronic infections of the brain, brain cancer, stroke rehabilitation, inborn errors of metabolism, autism, and mental retardation.

10. The composition of any one of claims 1-9 wherein the nanoconjugate has a mass that is at least about 400, about 600, about 800, about 1000, about 1200 or more Daltons.

11. The composition of any one of claims 1-10 wherein the nanoconjugate has a zeta potential measurement of from about -10 millivolts (mV) to about -50 millivolts (mV).

12. A method of treating a patient in need of a composition that is able to traverse the blood-brain barrier, comprising administering to the patient a therapeutically effective amount of a composition comprising a functionalized nanoconjugate, the nanoconjugate comprising a polynucleotide having a sequence sufficiently complementary to a target polynucleotide to hybridize to and inhibit expression of the target polynucleotide.

13. A method of administering a composition comprising a functionalized nanoconjugate to a patient, the method comprising administering to the patient a

therapeutically effective amount of the composition; wherein the nanoconjugate comprises a polynucleotide having a sequence sufficiently complementary to a target polynucleotide to hybridize to and inhibit expression of the target polynucleotide, said composition having the ability to traverse the blood-brain barrier, and wherein the patient is in need of a composition that is able to traverse the blood- brain barrier.

14. The method of claim 12 or claim 13 wherein the composition further comprises a therapeutic agent.

15. The method of any one of claims 12-14 wherein the patient suffers from a central nervous system (CNS) disorder.

16. The method of any one of claims 12-15 wherein the patient suffers from a disorder caused by aberrant gene expression.

17. The method of any one of claims 12-16 wherein the patient suffers from an acute and/or chronic disorder.

18. The method of claim 17 wherein the acute disorder is selected from the group consisting of focal brain ischemia, global brain ischemia, brain trauma, spinal cord injury, acute infections, status epilepticus, migraine headache, acute psychosis, suicidal depression, acute anxiety/phobia and injury related maladies.

19. The method of claim 17 wherein the chronic disorder is selected from the group consisting of chronic neurodegeneration, retinal degeneration, depression, chronic affective disorders, lysosmal storage disorders, chronic infections of the brain, brain cancer, stroke rehabilitation, inborn errors of metabolism, autism, and mental retardation.

20. The method of any one of claims 12-19 wherein the composition is administered only once.

21. The method of any one of claims 12-19 wherein the composition is administered at a frequency of no greater than about once per week.

22. The method of any one of claims 12-21 wherein the patient is a human.

Description:
NANOCONJUGATES ABLE TO CROSS THE BLOOD-BRAIN BARRIER CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the priority benefit under 35 U.S.C. § 119(e) of U.S.

Provisional Application No. 61/534,853, filed September 14, 2011, the disclosure of which is incorporated herein by reference in its entirety.

STATEMENT OF GOVERNMENT INTEREST

[0002] This invention was made with government support under Grant Number U54 CA151880 awarded by the National Institutes of Health/National Cancer Institute. The government has certain rights in the invention.

FIELD OF THE INVENTION

[0003] The present disclosure is directed to nanoconjugates that cross the blood-brain barrier and methods of their therapeutic use.

BACKGROUND OF THE INVENTION

[0004] The brain is unique in allowing only select access to molecules. While this is a useful protective mechanism, it also prevents potentially beneficial molecular agents from gaining access to the central nervous system (CNS), and as such, the molecular agents are unable to exert a therapeutic effect in many neurological disorders or other conditions of the CNS.

[0005] The blood-brain barrier (BBB) performs a neuroprotective function by tightly controlling access to the brain; consequently it also impedes access of pharmacological agents to cerebral tissues, necessitating the use of vectors for their transit. Blood-brain barrier (BBB) permeability is frequently a rate-limiting factor for the penetration of drugs or peptides into the CNS [Pardridge, Neurovirol. 5: 556-569 (1999); Bickel et al., Adv. Drug Deliv. Rev. 46: 247-279 (2001)]. The brain is shielded against potentially toxic substances by the BBB, which is formed by brain capillary endothelial cells that are closely sealed by tight junctions. In addition, brain capillaries possess few fenestrae and few endocytic vesicles, compared to the capillaries of other organs [Pardridge, Neurovirol. 5: 556-569 (1999)]. There is little transit across the BBB of large, hydrophilic molecules aside from some specific proteins such as transferrin, lactoferrin and low-density lipoproteins, which are taken up by receptor-mediated transcytosis (RMT) [Pardridge, Neurovirol. 5: 556-569 (1999); Tsuji et al., Adv. Drug Deliv. Rev. 36: 277-290 (1999); Kusuhara et al., Drug Discov. Today 6: 150-156 (2001); Dehouck et al. J. Cell. Biol. 138: 877-889 (1997); and Fillebeen et al., J. Biol. Chem. 274: 7011-7017 (1999)].

[0006] Malignant glioma (MG) represent the most prevalent and lethal primary cancer of the central nervous system. Patients diagnosed with the highest grade MG, grade IV glioblastoma multiforme (GBM), survive for only 9-12 months after diagnosis despite surgical resection and aggressive treatment regimens. Multimodal approaches using radiation with conjunctive chemotherapy (temozolamide (TMZ)) resulted in only marginal increase in patients' survival up to 14.6 months. Furthermore, recurrence is nearly universal and salvage therapies for such progression remain ineffective. GBM remains a highly enigmatic and incurable disease particularly due to a highly therapy-resistant cancer stem cell population (brain tumor stem cell, BTSC) and an incomplete understanding of how catalogued genetic aberrations dictate phenotypic hallmarks of the disease. It is highly resistant even to intense therapy (apoptosis) despite florid intratumoral necrogenesis. The continued lack of success in treating high-grade gliomas with targeted receptor tyrosine kinase inhibitors, which have been proven to be effective in other malignancies, has prompted a reevaluation of all aspects of glioma drug development and underlined the overarching need to develop an innovative technological platform and refine cell culture-based and in vivo model systems to combat the disease.

SUMMARY OF THE INVENTION

[0007] Polyvalent nanoconjugates address the critical challenges described above on multiple levels. The single-entity, targeted therapeutic is able to cross the blood-brain barrier (BBB) and is thus effective in the treatment of central nervous system (CNS) disorders. Further, despite the tremendously high cellular uptake of nanoconjugates, they exhibit no toxicity in the cell types tested thus far (see Table 1, below). This property is critical for therapeutic agent delivery applications for reducing off-target effects.

Cell Type Designation or Source

Breast SKBR3, MDA-MB-321, AU-565

Brain U87, LN229

Bladder HT- 1376, 5637, T24

Colon LS513

Cervix HeLa, SiHa

Skin C166, KB, MCF, 10A

Kidney MDCK

Blood Sup Tl, Jurkat

Leukemia K562

Liver HepG2

Kidney 293T

Ovary CHO

Macrophage RAW 264.7

Hippocampus Neurons primary, rat

Astrocytes primary, rat

Glial Cells primary, rat

Bladder primary, human

Erythrocytes primary, mouse

Peripheral Blood Mononuclear Cell primary, mouse

T-Cells primary, human

Beta Islets primary, mouse

Skin primary, mouse

Table 1

[0008] While some of the cell types shown in Table 1 are cells of the brain/nervous system, the data was gathered from in vitro experiments.

[0009] In one aspect, the disclosure provides a composition comprising a nanoconjugate, the nanoconjugate comprising a polynucleotide that is sufficiently complementary to a target polynucleotide which encodes a polypeptide specifically expressed in a central nervous system (CNS) disorder, the nanoconjugate having the ability to cross the blood-brain barrier (BBB). In some embodiments, the composition further comprises a targeting moiety. In various embodiments, the disorder is caused by aberrant gene expression. In some embodiments, the composition further comprises a therapeutic agent, and in further embodiments, the therapeutic agent is temozolamide. In some embodiments, the

nanoconjugate further comprises a targeting moiety and/or a therapeutic agent.

[0010] In further embodiments, it is contemplated that the disorder is acute and/or chronic.

[0011] In some embodiments, the acute disorder is selected from the group consisting of focal brain ischemia, global brain ischemia, brain trauma, spinal cord injury, acute infections, status epilepticus, migraine headache, acute psychosis, suicidal depression and acute anxiety/phobia, and injury related maladies, including but not limited to traumatic brain injury and swelling. In further embodiments, the chronic disorder is selected from the group consisting of chronic neurodegeneration, retinal degeneration, depression, chronic affective disorders, lysosmal storage disorders, chronic infections of the brain, brain cancer, stroke rehabilitation, inborn errors of metabolism, autism, and mental retardation.

[0012] In further embodiments, the nanoconjugate has a mass that is at least about 400, about 600, about 800, about 1000, about 1200 or more Daltons. In some embodiments, the nanoconjugate has a mass that is at least about 1, about 2, about 3, about 5, about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, about 100, about 200, about 500, about 700, about 900 or more kilodaltons.

[0013] In some embodiments, a nanoconjugate of the disclosure possesses a zeta potential (surface charge) measurement of from about -10 millivolts (mV) to about -50 millivolts (mV). In further embodiments, the nanoconjugate possesses a zeta potential measurement of from about -10 mV to about -40 mV, or from about -10 mV to about -30 mV, or from about - 20 mV to about -50 mV, or from about -20 mV to about -40 mV, or from about -30 mV to about -45 mV, or from about -30 mV to about -50 mV. In some embodiments, the nanoconjugate possesses a zeta potential measurement of about -10 mV, about -15 mV, about -20 mV, about -25 mV, about -30 mV, about -35 mV, about -40 mV, about -45 mV, about - 50 mV or about -60 mV.

[0014] In one aspect, the disclosure provides a method of treating a patient in need of a composition that is able to traverse the blood-brain barrier, comprising administering to the patient a therapeutically effective amount of a composition comprising a functionalized nanoconjugate, the nanoconjugate comprising a polynucleotide having a sequence sufficiently complementary to a target polynucleotide to hybridize to and inhibit expression of the target polynucleotide. In any of the aspects or embodiments of the disclosure, it is contemplated that the patient is a human.

[0015] In another aspect, the disclosure provides a method of administering a composition comprising a functionalized nanoconjugate to a patient, the method comprising administering to the patient a therapeutically effective amount of the composition; wherein the

nanoconjugate comprises a polynucleotide having a sequence sufficiently complementary to a target polynucleotide to hybridize to and inhibit expression of the target polynucleotide, said composition having the ability to traverse the blood-brain barrier, and wherein the patient is in need of a composition that is able to traverse the blood-brain barrier.

[0016] In any of the aspects or embodiments of the disclosure, a composition as described herein further comprises a therapeutic agent. In any of the aspects or embodiments of the disclosure, the patient suffers from a central nervous system (CNS) disorder. In any of the aspects or embodiments of the disclosure, the patient suffers from a disorder caused by aberrant gene expression.

[0017] In some embodiments, the patient suffers from an acute and/or chronic disorder. In embodiments where the patient suffers from an acute disorder, it is further contemplated that the acute disorder is selected from the group consisting of focal brain ischemia, global brain ischemia, brain trauma, spinal cord injury, acute infections, status epilepticus (SE), migraine headache, acute psychosis, suicidal depression and acute anxiety/phobia, and injury related maladies, including but not limited to traumatic brain injury and swelling.

[0018] In embodiments where the patient suffers from a chronic disorder, it is further contemplated that the chronic disorder is selected from the group consisting of chronic neurodegeneration, retinal degeneration, depression, chronic affective disorders, lysosmal storage disorders, chronic infections of the brain, brain cancer, stroke rehabilitation, inborn errors of metabolism, autism, and mental retardation.

[0019] In some embodiments, a composition of the disclosure is administered only once. In some embodiments, a composition of the disclosure is administered at a frequency of no greater than about once per week.

[0020] In another aspect, the disclosure provides a package or kit comprising (a) a nanoconjugate or composition comprising a nanoconjugate, optionally in a container, (b) optionally an additional therapeutic agent; and (c) a package insert, package label, instructions or other labeling directing or disclosing any of the methods or embodiments disclosed herein.

BRIEF DESCRIPTION OF THE DRAWINGS

[0021] Figure 1 depicts Bcl2L12 expression (A) and RTK activation profile (B) in selected glioma, BTSC lines and explants.

[0022] Figure 2 shows that RNA-nanoconjugates effectively silence Bcl2L12 expression. (A and B) Glioma cell lines and huBTSC_18 were treated with indicated amounts of RNA- nanoconjugates (control (Co)/scrambled-RNA sequence-RNA-Au NPs— Co-RNA- nanoconjugate and Bcl2L12 targeting RNA-nanoconjugates— L12-1- and L12-2-RNA- nanoconjugates) and subjected to qRT-PCR (A) and western blot (B) analyses. The migration positions of Bcl2L12 and Hsp70 are indicated. *-labeled band represents post- translationally modified Bcl2L12. (C) Knockdown efficacies were compared to

conventional lipoplex-delivered siRNAs. (D) For studies of knockdown persistence, LN235 cells were treated with L12-RNA-nanoconjugates (1 nM) for 5 days, and subjected to anti- Bcl2L12 western blot analyses. (E) RNA-nanoconjugate-mediated knockdown of Bcl2L12 results in enhanced caspase activation. Co-, L12-1- and L12-2- nanoconjugate-treated LN235 cells were treated with staurosporine (STS, 500 nM) for the indicated periods of time, lysed and subjected to western blot analyses for active caspases 3 and 7. The migration positions of the active subunits (large, LS; large and large+N peptide, LS+N) and Hsp70 (loading control) are indicated. Histograms quantify Bcl2L12 expression as assessed by densitometric analyses of corresponding western blots.

[0023] Figure 3 shows neutralization of aB-crystallin in LN235 cells results in reduced invasive potential and increases susceptibility towards STS-instigated apoptosis. (A) LN235 cells were treated with aB-crystallin-targeting nanoconjugates or siRNAs, lysed and subjected to western blot analyses using aB-crystallin and Hsp70-specific antibodies. (B) CRYAB- and Co-RNA-nanoconjugate-treated cells were subjected to Matrigel invasion assays and numbers of invading cells were quantified by trypan blue staining. (C) Co- CRYAB-1- and CRYAB-2-RNA-nanoconjugate-treated LN235 cells were treated and analyzed as described in Figure 2E. LS, large subunit; LS+N, large subunit+N peptide. Histograms quantify Bcl2L12 expression as assessed by densitometric analyses of corresponding western blots.

[0024] Figure 4 shows intratumoral uptake of nanoconjugates as assessed by confocal immunofluorescence of Cy5-labeled Au-NPs (A), by ICP-MS (C) and by MRI (D) of locally delivered DNA-Gd(III)-nanoconjugates in normal brain and explant structures. (B) shows quantification of dispersion over time see using confocal IF images of serial coronal sections.

[0025] Figure 5 shows intratumoral uptake of nanoconjugates— Intracranial versus Intravenous Administration as assessed by ICP-MS (A) Direct intracranial (LC.) delivery of Co-RNA-nanoconjugates (5μ1, 300nM). Nanoconjugates were locally delivered once and then the brain and tumor tissues were harvested 48 hours post administration. (B) Systemic intravenous (I.V.) injection of Co-RNA-nanoconjugates (100 μΐ, 300nM). Four injections of nanoconjugates every 48 hours. The tissues were harvested 24 hours after the fourth injection.

[0026] Figure 6 shows the effects of RNA-nanoconjugates on survival of tumor mice. Co- , L12-1-, and L12-2-RNA-nanoconjugates were administered to mice via tail vein injection (I.V.). Each mouse received 5 injections (1.4 mg/kg RNA per injection totaling at 7 mg/kg treatment, approximately 150-200 μ ΐ of RNA-nanoconjugates at 500 nM). No difference in survival days was seen between Co- and L12-1-RNA nanoconjugate treatment groups (p = 0.50). However, a significant difference was observed between Co- and L12-2-RNA- nanoconjugate treatment groups (p = 0.01).

DETAILED DESCRIPTION OF THE INVENTION

[0027] The blood brain barrier is a limiting factor in the delivery of many peripherally- administered agents to the central nervous system. The present disclosure provides nanoconjugates that are able to cross the BBB, and retain their activity once across the BBB. Various aspects of the invention address these factors, by providing nanoconjugates that have one or more biomolecules associated therewith. In some embodiments, the nanoconjugate is further associated or co-administered with a therapeutic agent.

[0028] "Treatment" or "treating" as used herein includes achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder or condition being treated. For example, in an individual with a neurological disorder, therapeutic benefit includes partial or complete halting of the progression of the disorder, or partial or complete reversal of the disorder. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological or psychological symptoms associated with the underlying condition such that an improvement is observed in the patient, notwithstanding the fact that the patient may still be affected by the condition. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition (e.g. , slowing the progression of a

neurological disorder), or decreasing the likelihood of occurrence of a condition. As used herein, "treating" or "treatment" includes prophylaxis.

[0029] In some embodiments, a nanoconjugate is "administered peripherally" or

"peripherally administered." As used herein, these terms refer to any form of administration of a nanoconjugate, optionally co-administered with a therapeutic agent, to an individual that is not direct administration to the CNS, i.e. , that brings the agent in contact with the non-brain side of the blood-brain barrier. "Peripheral administration," as used herein, includes intravenous, subcutaneous, intramuscular, intraperitoneal, transdermal, inhalation, transbuccal, intranasal, rectal, and oral administration.

[0030] As used herein, "hybridization" means an interaction between two or three strands of nucleic acids by hydrogen bonds in accordance with the rules of Watson-Crick DNA complementarity, Hoogstein binding, or other sequence- specific binding known in the art. Hybridization can be performed under different stringency conditions known in the art. "Specifically hybridize," as used herein, is hybridization that allows for a stabilized duplex between polynucleotide strands that are complementary or substantially complementary. For example, a polynucleotide strand having 21 nucleotide units can base pair with another polynucleotide of 21 nucleotide units, yet only 19 bases on each strand are complementary or sufficiently complementary, such that the "duplex" has 19 base pairs. The remaining bases may, for example, exist as 5' and/or 3' overhangs. Further, within the duplex, 100% complementarity is not required; substantial complementarity is allowable within a duplex. Sufficient complementarity refers to 75% or greater complementarity. For example, a mismatch in a duplex consisting of 19 base pairs results in 94.7% complementarity, rendering the duplex sufficiently complementary.

[0031] The terms "therapeutically effective amount," as used herein, refer to an amount of a compound sufficient to treat, ameliorate, or prevent the identified disease or condition, or to exhibit a detectable therapeutic, prophylactic, or inhibitory effect. The effect can be detected by, for example, an improvement in clinical condition, or reduction in symptoms. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Where a drug has been approved by the U.S. Food and Drug Administration (FDA), a "therapeutically effective amount" refers to the dosage approved by the FDA or its counterpart foreign agency for treatment of the identified disease or condition.

[0032] As used herein, a patient "in need of a composition that is able to traverse the blood-brain barrier " is a patient who would benefit from a composition that is able to traverse the blood-brain barrier. The patient may be suffering from any disease or condition for which therapy with a composition that is able to traverse the blood-brain barrier may be useful in ameliorating symptoms.

[0033] A "disorder of the CNS" or "CNS disorder," as those terms are used herein, encompasses any condition that affects the brain and/or spinal cord and that leads to suboptimal function. In some embodiments, the disorder is an acute disorder. Acute disorders of the CNS include focal brain ischemia, global brain ischemia, brain trauma, spinal cord injury, acute infections, status epilepticus (SE), migraine headache, acute psychosis, suicidal depression, and acute anxiety/phobia. In some embodiments, the disorder is a chronic disorder. Chronic disorders of the CNS include chronic neurodegeneration, retinal degeneration, depression, chronic affective disorders, lysosmal storage disorders, chronic infections of the brain, brain cancer, stroke rehabilitation, inborn errors of metabolism, autism, mental retardation. Chronic neurodegeneration includes neurodegenerative diseases such as prion diseases, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), transverse myelitis, motor neuron disease, Pick's disease, tuberous sclerosis, lysosomal storage disorders, Canavan's disease, Rett's syndrome, spinocerebellar ataxias, Friedreich's ataxia, optic atrophy, and retinal degeneration, and aging of the CNS.

[0034] As used herein, "concomitant use" is understood to be interchangeable with concurrent administration or co-administration. Thus, the terms are understood to encompass administration simultaneously, or at different times, and by the same route or by different routes, as long as the two agents are given in a manner that allows both agents to be affecting the body at the same time. For example, concomitant use can refer to a medication concomitantly administered, whether prescribed by the same or a different practitioner, or for the same or a different indication.

[0035] It is noted here that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise.

[0036] It is also noted that the term "about" as used herein is understood to mean approximately.

[0037] It is further noted that the terms "attached," "conjugated" and "functionalized" are used interchangeably herein and refer to the association of a polynucleotide, peptide, polypeptide, therapeutic agent, contrast agent and a combination thereof with a

nanoconjugate.

THE BLOOD-BRAIN BARRIER (BBB)

[0038] As used herein, the "blood-brain barrier" (BBB) refers to the barrier between the peripheral circulation and the brain and spinal cord which is formed by tight junctions within the brain capillary endothelial plasma membranes, that creates a highly selective barrier that restricts the transport of molecules into the brain. The blood-brain barrier within the brain, the blood-spinal cord barrier within the spinal cord, and the blood-retinal barrier within the retina, are contiguous capillary barriers within the central nervous system (CNS), and are collectively referred to as the blood-brain barrier or BBB.

[0039] The BBB is formed by epithelial-like high resistance tight junctions within the endothelium of capillaries perfusing the vertebrate brain. Unless a therapeutic molecule is lipid-soluble with a molecular weight of 400-600 Daltons or less, brain penetration is limited [Pardridge, Curr Opin Pharmacol 6: 494-500 (2006)]. Because of the presence of the BBB, circulating molecules gain access to brain cells only via one of two processes: (i) lipid- mediated transport of small molecules through the BBB by free diffusion, or (ii) catalyzed transport. The latter includes carrier-mediated transport processes for low molecular weight nutrients and water soluble vitamins or receptor-mediated transport for circulating peptides (e.g., insulin), plasma proteins (e.g., transferrin), or viruses. While BBB permeability, per se, is controlled by the biochemical properties of the plasma membranes of the capillary endothelial cells, overall brain microvascular biology is a function of the paracrine interactions between the capillary endothelium and the other two major cells comprising the microcirculation of brain, i.e., the capillary pericyte, which shares the basement membrane with the endothelial cell, and the astrocyte foot process, which invests 99% of the abluminal surface of the capillary basement membrane in brain. Microvascular functions frequently ascribed to the capillary endothelium are actually executed by either the capillary pericyte or the capillary astrocyte foot process [Pardridge, J. Neurovir. 5: 556-569 (1999)].

[0040] The BBB largely defines the operating environment of the CNS by regulating the movement of substances between the blood and the CSF and brain interstitial fluid. The BBB is often divided into the vascular, or endothelial, barrier and the epithelial barrier at the choroid plexus (also termed the blood-CSF barrier). The endothelial cells that comprise the capillaries and line the arterioles and venules constitute the barrier function of the spinal cord and in most areas of the brain [Rapoport, Blood Brain Barrier in Physiology and Medicine, Raven Press, New York. (1976)]. The endothelial cells are modified in that circumferential belts of tight junctions between contiguous non-fenestrated endothelial cells of the CNS preclude the leakage found in the capillary beds of peripheral tissues. Intracellular tight junctions comparable to those of the brain endothelium exist between contiguous epithelial cells at the choroid plexus [Johanson, The choroid plexus-arachnoid membrane-cerebro spinal fluid system. In: Neuronal Microenvironment. Boulton AA, Baker GB, Walz W(eds). The Humana Press: Clifton, New Jersey, pp 33-104 (1988)] and between arachnoid mater cells [Balin et al., J Comp Neurol 251: 260-280 (1986)]. The brain endothelia have other modifications as well. They do engage in endocytosis of blood-borne macromolecules and a recycling of the luminal plasmalemma but to a lesser degree than peripheral endothelia and choroid plexus [Broadwell et al., Cell biological perspective for the transcytosis of peptides and proteins through the mammalian blood-brain fluid barriers. In: The Blood-Brain Barrier. Pardridge WM (ed). Raven Press Ltd: New York, pp 165-199 (1993)]. Secondary lysosomes hydrolyze many but not all macromolecules undergoing endocytosis within the BBB endothelia [Broadwell et al., Proc Natl. Acad Sci. USA 78: 7820-7824 (1981);

Broadwell et al., Cell biological perspective for the transcytosis of peptides and proteins through the mammalian blood-brain fluid barriers. In: The Blood-Brain Barrier. Pardridge WM (ed). Raven Press Ltd: New York, pp 165-199 (1993)]. These modifications of the endothelia effectively eliminate the plasma ultrafiltrate characteristic of capillary beds in peripheral tissues and serve to define the restrictive permeability of the BBB [Banks, J.

Neurovir. 5: 538-555 (1999)].

[0041] Therefore, most potentially therapeutic, diagnostic, or research molecules do not cross the BBB in pharmacologically active amounts. So as to bypass the BBB, invasive transcranial drug delivery strategies are used, such as intracerebro -ventricular (ICV) infusion, intracerebral (IC) administration, and convection enhanced diffusion (CED). Transcranial drug delivery to the brain is expensive, invasive, and largely ineffective. The ICV route typically delivers drugs only to the ependymal surface of the brain, not into brain

parenchyma. The IC administration of a neurotrophin, such as nerve growth factor (NGF), only delivers drug to the local injection site, owing to the low efficiency of drug diffusion within the brain. The CED of neurotrophin results in preferential fluid flow through the white matter tracts of brain, which causes demyelination, and astrogliosis.

[0042] The present disclosure offers an alternative to these highly invasive and generally unsatisfactory methods for bypassing the BBB, allowing nanoconjugates to cross the BBB from the peripheral blood. It is based on the use of nanoconjugates that are able to transport a desired substance from the peripheral blood to the CNS. Given that brain penetration of a therapeutic agent that has a mass of 400-600 Daltons or more is limited, and further that the mass of a single base of DNA is approximately 320 daltons, it is unexpected that data presented herein demonstrates that a nanoconjugate that is functionalized with a multitude of polynucleotides, each comprising a multitude of bases, is able to cross the BBB in any appreciable quantity.

[0043] In one aspect, the disclosure provides compositions and methods that utilize a nanoconjugate capable of crossing the BBB. The compositions and methods are useful in transporting nanoconjugates and, optionally, a therapeutic agent, from the peripheral blood and across the blood brain barrier into the CNS.

NANOCONJUGATES

[0044] The compositions of the disclosure comprise a nanoconjugate. A nanoconjugate comprises a nanoparticle that is, in certain aspects, hollow. Nanoconjugates further comprise, in various embodiments, a biomolecule. As used herein, a "biomolecule" is understood to include a polynucleotide, peptide, polypeptide, small molecule, therapeutic agent, contrast agent, and a combination thereof. In various aspects of the nanoconjugate, all of the biomolecules are identical, or in the alternative, at least two biomolecules are different.

[0045] The nanoconjugates of the disclosure are not polymer-based nanoconjugates. Thus, nanoconjugates comprising, for example, polyethylene glycol (PEG)-coated

hexadecylcyanoarcylate nanospheres; poly(butylcyanoacrylate) nanoparticles;

poly(butylcyanoacrylate) nanoparticles coated with polysorbate 80; lipid nanoparticles; lipid nanoparticles consisting of emulsions of solidified oil nanodroplets loaded with, for example, iron oxide; or a nanogel consisting of cross-linked PEG and polyethylenimine are not contemplated aspects or embodiments of this disclosure.

[0046] The disclosure provides nanoconjugates that, in various embodiments, have a mass that is at least about 400, about 600, about 800, about 1000, about 1200 or more Daltons. In further embodiments, the nanoconjugate has a mass that is at least about 1, about 2, about 3, about 5, about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, about 100, about 200, about 500, about 700, about 900 or more kilodaltons. As used herein, the mass of a nanoconjugate is understood to include the mass of a nanoparticle (if present) plus the mass of any biomolecules and/or therapeutic agent(s) that are associated with the nanoparticle.

Nanoparticle

[0047] Nanoparticles are provided which are functionalized, in some aspects, to have a biomolecule attached thereto. The size, shape and chemical composition of the nanoparticles contribute to the properties of the resulting functionalized nanoparticle. These properties include for example, optical properties, optoelectronic properties, electrochemical properties, electronic properties, stability in various solutions, magnetic properties, and pore and channel size variation. Mixtures of nanoparticles having different sizes, shapes and/or chemical compositions, as well as the use of nanoparticles having uniform sizes, shapes and chemical composition, and therefore a mixture of properties are contemplated. Examples of suitable particles include, without limitation, aggregate particles, isotropic (such as spherical particles), anisotropic particles (such as non-spherical rods, tetrahedral, and/or prisms) and core-shell particles, such as those described in U.S. Patent No. 7,238,472 and International Publication No. WO 2003/08539, the disclosures of which are incorporated by reference in their entirety.

[0048] In one embodiment, the nanoparticle is metallic, and in various aspects, the nanoparticle is a colloidal metal. Thus, in various embodiments, nanoparticles of the invention include metal (including for example and without limitation, silver, gold, platinum, aluminum, palladium, copper, cobalt, indium, nickel, or any other metal amenable to nanoparticle formation), semiconductor (including for example and without limitation, CdSe, CdS, and CdS or CdSe coated with ZnS) and magnetic (for example, ferromagnetite) colloidal materials.

[0049] Also, as described in U.S. Patent Publication No 2003/0147966, nanoparticles of the invention include those that are available commercially, as well as those that are synthesized, e.g., produced from progressive nucleation in solution (e.g., by colloid reaction) or by various physical and chemical vapor deposition processes, such as sputter deposition. See, e.g., HaVashi, Vac. Sci. Technol. A5(4) : 1375-84 (1987); Hayashi, Physics Today, 44- 60 (1987); MRS Bulletin, January 1990, 16-47. As further described in U.S. Patent

Publication No 2003/0147966, nanoparticles contemplated are alternatively produced using HAuCl 4 and a citrate-reducing agent, using methods known in the art. See, e.g., Marinakos et al, Adv. Mater. 11:34-37(1999); Marinakos et al, Chem. Mater. 10: 1214-19(1998); Enustun & Turkevich, J. Am. Chem. Soc. 85: 3317(1963).

[0050] Nanoparticles can range in size from about 1 nm to about 250 nm in mean diameter, about 1 nm to about 240 nm in mean diameter, about 1 nm to about 230 nm in mean diameter, about 1 nm to about 220 nm in mean diameter, about 1 nm to about 210 nm in mean diameter, about 1 nm to about 200 nm in mean diameter, about 1 nm to about 190 nm in mean diameter, about 1 nm to about 180 nm in mean diameter, about 1 nm to about 170 nm in mean diameter, about 1 nm to about 160 nm in mean diameter, about 1 nm to about 150 nm in mean diameter, about 1 nm to about 140 nm in mean diameter, about 1 nm to about 130 nm in mean diameter, about 1 nm to about 120 nm in mean diameter, about 1 nm to about 110 nm in mean diameter, about 1 nm to about 100 nm in mean diameter, about 1 nm to about 90 nm in mean diameter, about 1 nm to about 80 nm in mean diameter, about 1 nm to about 70 nm in mean diameter, about 1 nm to about 60 nm in mean diameter, about 1 nm to about 50 nm in mean diameter, about 1 nm to about 40 nm in mean diameter, about 1 nm to about 30 nm in mean diameter, or about 1 nm to about 20 nm in mean diameter, about 1 nm to about 10 nm in mean diameter. In other aspects, the size of the nanoparticles is from about 5 nm to about 150 nm (mean diameter), from about 5 to about 50 nm, from about 10 to about 30 nm, from about 10 to 150 nm, from about 10 to about 100 nm, or about 10 to about 50 nm. The size of the nanoparticles is from about 5 nm to about 150 nm (mean diameter), from about 30 to about 100 nm, from about 40 to about 80 nm. The size of the nanoparticles used in a method varies as required by their particular use or application. The variation of size is advantageously used to optimize certain physical characteristics of the nanoparticles, for example, optical properties or the amount of surface area that can be functionalized as described herein.

[0051] Nanoparticles of larger diameter are, in some aspects, contemplated to be functionalized with a greater number of biomolecules [Hurst et al. , Analytical Chemistry 78(24): 8313-8318 (2006)] during nanoconjugate production. In some aspects, therefore, the number of biomolecules used in the production of a nanoconjugate is from about 10 to about 25,000 biomolecules per nanoconjugate. In further aspects, the number of biomolecules used in the production of a nanoconjugate is from about 50 to about 10,000 biomolecules per nanoconjugate, or from about 200 to about 5,000 biomolecules per nanoconjugate, or from about 50 to about 100 biomolecules per nanoconjugate, or from about 20 to about 100 biomolecules per nanoconjugate, or from about 20 to about 50 biomolecules per

nanoconjugate. Thus, in various embodiments, the number of biomolecules used in the production of a nanoconjugate is about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 500, about 600, about 700, about 800, about 900, about 1,000, about 2,000, about 5000, about 10,000, about 15,000, about 20,000, about 25,000 or more per nanoconjugate. Hollow nanoconjugates

[0052] As described herein, in various aspects the nanoconjugates provided by the disclosure are hollow. The porosity and/or rigidity of a hollow nanoconjugate depends in part on the density of biomolecules that are crosslinked on the surface of a nanoparticle during nanoconjugate production. In general, a lower density of biomolecules crosslinked on the surface of the nanoparticle results in a more porous nanoconjugate, while a higher density of biomolecules crosslinked on the surface of the nanoparticle results in a more rigid nanoconjugate. Porosity and density of a hollow nanoconjugate also depends on the degree and type of crosslinking between biomolecules.

[0053] Methods of making hollow nanoconjugates are known in the art, and are generally described in International Patent Application Number PCT/US2010/055018 and Zhang et al. [J Am Chem Soc. 132(43): 15151-15153 (2010)], which are each incorporated by reference herein in their entirety.

[0054] In some embodiments, hollow nanoconjugates are prepared via poly alkyne chemistry [Zhang et al, J Am Chem Soc. 132(43): 15151-15153 (2010)]. Additional cross linking strategies, such as through the use of a homobifunctional cross linker {e.g., Sulfo- EGS) or other reactive group (for example and without limitation, amines, amides, alcohols, esters, aldehydes, ketones, thiols, disulfides, carboxylic acids, phenols, imidazoles, hydrazines, hydrazones, azides, and alkynes) are also contemplated.

[0055] An additional method of preparing a hollow nanoconjugate, called surface assisted crosslinking (SAC), comprises a mixed monolayer of modified nucleic acids and reactive thiolated molecules that are assembled on the nanoparticle surface and crosslinked together. As used herein, a "monolayer" means that only a single stratum of biomolecules is crosslinked at the surface of a nanoconjugate. A biomolecule as used herein includes without limitation a polynucleotide, peptide, polypeptide, small molecule, therapeutic agent, contrast agent and a combination thereof.

[0056] The chemical that causes crosslinking of the biomolecules of interest are known to those of skill in the art, and include without limitation Disuccinimidyl glutarate,

Disuccinimidyl suberate, Bis[sulfosuccinimidyl] suberate, Tris-succinimidyl aminotriacetate, succinimidyl 4-hydrazinonicotinate acetone hydrazone, succinimidyl 4- hydrazidoterephthalate hydrochloride, succinimidyl 4-formylbenzoate,

Dithiobis[succinimidyl propionate], 3,3'-Dithiobis[sulfosuccinimidylpropionate], Disuccinimidyl tartarate, Bis[2-(succinimidooxycarbonyloxy)ethyl]sulfone, Ethylene glycol bis[succinimidylsuccinate], Ethylene glycol bis[sulfosuccinimidylsuccinate], Dimethyl adipimidate » 2 HC1, Dimethyl pimelimidate » 2 HC1, Dimethyl Suberimidate » 2 HC1, 1,5- Difluoro-2,4-dinitrobenzene, B-[Tris(hydroxymethyl) phosphino] propionic acid, Bis- Maleimidoethane, 1,4-bismaleimidobutane, Bismaleimidohexane, Tris[2- maleimidoethyl] amine, 1,8-Bis-maleimido-diethyleneglycol, 1,11-Bis-maleimido- triethyleneglycol, 1,4 bismaleimidyl-2,3-dihydroxybutane, Dithio-bismaleimidoethane, 1,4- Di-[3'-(2'-pyridyldithio)-propionamido]butane, 1,6-Hexane-bis-vinylsulfone, Bis-[b-(4- Azidosalicylamido)ethyl]disulfide, N-(a-Maleimidoacetoxy) succinimide ester, N-[B- Maleimidopropyloxy]succinimide ester, N-[g-Maleimidobutyryloxy] succinimide ester, N-[g- Maleimidobutyryloxy] sulfo succinimide ester, m-Maleimidobenzoyl-N-hydroxysuccinimide ester, m-Maleimidobenzoyl-N-hydroxysulfosuccinimide ester, Succinimidyl 4-[N- maleimidomethyl]cyclohexane-l-carboxylate, Sulfosuccinimidyl 4-[N- maleimidomethyl]cyclohexane-l-carboxylate, N-e-Maleimidocaproyloxy] succinimide ester, N-e-Maleimidocaproyloxy]sulfosuccinimide ester, Succinimidyl 4-[p- maleimidophenyl]butyrate, Sulfosuccinimidyl 4-[p-maleimidophenyl]butyrate, Succinimidyl-

6-[B-maleimidopropionamido]hexanoate, Succinimidyl-4-[N-Maleimidomethyl]cyclohexane- l-carboxy-[6-amidocaproate], N-[k-Maleimidoundecanoyloxy]sulfosuccinimide ester, N -

Succinimidyl 3-(2-pyridyldithio)-propionate, Succinimidyl 6-(3-[2-pyridyldithio]- propionamido)hexanoate, 4-Succinimidyloxycarbonyl-methyl-a- [2-pyridyldithio]toluene, 4-

Sulfosuccinimidyl-6-methyl-a-(2-pyridyldithio)toluamido]h exanoate), N-Succinimidyl iodoacetate, Succinimidyl 3-[bromoacetamido]propionate, N-Succinimidyl[4- iodoacetyl]aminobenzoate, N-Sulfosuccinimidyl[4-iodoacetyl]aminobenzoate, N-

Hydroxysuccinimidyl-4-azidosalicylic acid, N-5-Azido-2-nitrobenzoyloxysuccinimide, N-

Hydroxysulfosuccinimidyl-4-azidobenzoate, Sulfosuccinimidyl[4-azidosalicylamido]- hexanoate, N-Succinimidyl-6-(4'-azido-2'-nitrophenylamino) hexanoate, N-

Sulfosuccinimidyl-6-(4'-azido-2'-nitrophenylamino) hexanoate, Sulfosuccinimidyl-

(perfluoroazidobenzamido)-ethyl- 1 ,3'-dithioproprionate, Sulfosuccinimidyl-2-(m-azido-o- nitrobenzamido)-ethyl-l,3'-proprionate, Sulfosuccinimidyl 2-[7-amino-4-methylcoumarin-3- acetamido]ethyl-l,3'dithiopropionate, Succinimidyl 4,4'-azipentanoate, Succinimidyl 6-(4,4'- azipentanamido)hexanoate , Succinimidyl 2-([4,4'-azipentanamido]ethyl)-l,3'- dithioproprionate , Sulfosuccinimidyl 4,4'-azipentanoate , Sulfosuccinimidyl 6-(4,4'- azipentanamido)hexanoate, Sulfosuccinimidyl 2-([4,4'-azipentanamido]ethyl)- 1 ,3'- dithioproprionate, Dicyclohexylcarbodiimide, l-Ethyl-3-[3- dimethylaminopropyl]carbodiimide Hydrochloride, N-[4-(p-Azidosalicylamido) butyl]-3'- (2'-pyridyldithio)propionamide, N-[B-Maleimidopropionic acid] hydrazide, trifluoroacetic acid salt, [N-e-Maleimidocaproic acid] hydrazide, trifluoroacetic acid salt, 4-(4-N- Maleimidophenyl)butyric acid hydrazide hydrochloride, N-[k-Maleimidoundecanoic acid]hydrazide, 3-(2-Pyridyldithio)propionyl hydrazide, p-Azidobenzoyl hydrazide, N-[p- Maleimidophenyl]isocyanate, and Succinimidyl-[4-(psoralen-8-yloxy)]-butyrate.

BIOMOLECULES

[0057] As described herein, a biomolecule includes without limitation a polynucleotide, peptide, polypeptide, small molecule, therapeutic agent, contrast agent and a combination thereof. In various aspects of the disclosure a biomolecule as described herein is covalently associated with the nanoparticle.

Polynucleotides

[0058] Polynucleotides contemplated by the present disclosure include DNA, RNA, modified forms and combinations thereof as defined herein. Accordingly, in some aspects, the nanoconjugate comprises DNA. In some embodiments, the DNA is double stranded, and in further embodiments the DNA is single stranded. In further aspects, the nanoconjugate comprises RNA, and in still further aspects the nanoconjugate comprises double stranded RNA, and in a specific embodiment, the double stranded RNA agent is a small interfering RNA (siRNA). The term "RNA" includes duplexes of two separate strands, as well as single stranded structures. Single stranded RNA also includes RNA with secondary structure. In one aspect, RNA having a hairpin loop is contemplated.

[0059] When a nanoconjugate comprise a plurality of structural polynucleotides, the polynucleotide is, in some aspects, comprised of a sequence that is sufficiently

complementary to a target sequence of a polynucleotide such that hybridization of the polynucleotide that is part of the nanoconjugate and the target polynucleotide takes place. The polynucleotide in various aspects is single stranded or double stranded, as long as the double stranded molecule also includes a single strand sequence that hybridizes to a single strand sequence of the target polynucleotide. In some aspects, hybridization of the polynucleotide that is part of the nanoconjugate can form a triplex structure with a double- stranded target polynucleotide. In another aspect, a triplex structure can be formed by hybridization of a double-stranded polynucleotide that is part of a nanoconjugate to a single- stranded target polynucleotide. Further description of triplex polynucleotide complexes is found in PCT/US2006/40124, which is incorporated herein by reference in its entirety.

[0060] In some aspects, polynucleotides contain a spacer as described herein. The spacer, in one aspect, comprises one or more crosslinking moieties that facilitate the crosslinking of one polynucleotide to another polynucleotide.

[0061] A "polynucleotide" is understood in the art to comprise individually polymerized nucleotide subunits. The term "nucleotide" or its plural as used herein is interchangeable with modified forms as discussed herein and otherwise known in the art. In certain instances, the art uses the term "nucleobase" which embraces naturally- occurring nucleotide, and non- naturally- occurring nucleotides which include modified nucleotides. Thus, nucleotide or nucleobase means the naturally occurring nucleobases adenine (A), guanine (G), cytosine (C), thymine (T) and uracil (U). Non-naturally occurring nucleobases include, for example and without limitations, xanthine, diaminopurine, 8-oxo-N6-methyladenine, 7-deazaxanthine, 7-deazaguanine, N4,N4-ethanocytosin, N',N'-ethano-2,6-diaminopurine, 5-methylcytosine (mC), 5-(C3— C6)-alkynyl-cytosine, 5-fluorouracil, 5-bromouracil, pseudoisocytosine, 2- hydroxy-5-methyl-4-tr- iazolopyridin, isocytosine, isoguanine, inosine and the "non-naturally occurring" nucleobases described in Benner et ah, U.S. Pat. No. 5,432,272 and Susan M. Freier and Karl-Heinz Altmann, 1997, Nucleic Acids Research, vol. 25: pp 4429-4443. The term "nucleobase" also includes not only the known purine and pyrimidine heterocycles, but also heterocyclic analogues and tautomers thereof. Further naturally and non-naturally occurring nucleobases include those disclosed in U.S. Pat. No. 3,687,808 (Merigan, et ah), in Chapter 15 by Sanghvi, in Antisense Research and Application, Ed. S. T. Crooke and B. Lebleu, CRC Press, 1993, in Englisch et ah, 1991, Angewandte Chemie, International Edition, 30: 613-722 (see especially pages 622 and 623, and in the Concise Encyclopedia of Polymer Science and Engineering, J. I. Kroschwitz Ed., John Wiley & Sons, 1990, pages 858-859, Cook, Anti-Cancer Drug Design 1991, 6, 585-607, each of which are hereby incorporated by reference in their entirety). In various aspects, polynucleotides also include one or more "nucleosidic bases" or "base units" which are a category of non-naturally- occurring nucleotides that include compounds such as heterocyclic compounds that can serve like nucleobases, including certain "universal bases" that are not nucleosidic bases in the most classical sense but serve as nucleosidic bases. Universal bases include 3-nitropyrrole, optionally substituted indoles {e.g., 5-nitroindole), and optionally substituted hypoxanthine. Other desirable universal bases include, pyrrole, diazole or triazole derivatives, including those universal bases known in the art.

[0062] Modified nucleotides are described in EP 1 072 679 and WO 97/12896, the disclosures of which are incorporated herein by reference. Modified nucleotides include without limitation, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and

2- thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and

3- deazaadenine. Further modified bases include tricyclic pyrimidines such as phenoxazine cytidine(lH-pyrimido[5 ,4-b] [l,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H- pyrimido[5 ,4-b] [l,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido[5,4-b] [l,4]benzox- azin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H- pyrido[3',2':4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified bases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deazaadenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Additional nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et ah , 1991, Angewandte Chemie, International Edition, 30: 613, and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and

Applications, pages 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press, 1993. Certain of these bases are useful for increasing the binding affinity and include 5-substituted

pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2- aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine

substitutions have been shown to increase nucleic acid duplex stability by 0.6- 1.2° C and are, in certain aspects combined with 2'-0-methoxyethyl sugar modifications. See, U.S. Pat. Nos. 3,687,808, U.S. Pat. Nos. 4,845,205; 5, 130,302; 5,134,066; 5, 175,273; 5,367,066; 5,432,272; 5,457, 187; 5,459,255; 5,484,908; 5,502,177; 5,525,711 ; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,830,653; 5,763,588; 6,005,096; 5,750,692 and 5,681,941, the disclosures of which are incorporated herein by reference.

[0063] Methods of making polynucleotides of a predetermined sequence are well-known. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd ed. 1989) and F. Eckstein (ed.) Oligonucleotides and Analogues, 1st Ed. (Oxford University Press, New York, 1991). Solid-phase synthesis methods are preferred for both polyribonucleotides and polydeoxyribonucleotides (the well-known methods of synthesizing DNA are also useful for synthesizing RNA). Polyribonucleotides can also be prepared enzymatically. Non-naturally occurring nucleobases can be incorporated into the polynucleotide, as well. See, e.g., U.S. Patent No. 7,223,833; Katz, J. Am. Chem. Soc, 74:2238 (1951); Yamane, et al, J. Am.

Chem. Soc, 83:2599 (1961); Kosturko, et al, Biochemistry, 13:3949 (1974); Thomas, J. Am. Chem. Soc, 76:6032 (1954); Zhang, et al, J. Am. Chem. Soc, 127:74-75 (2005); and Zimmermann, et al., J. Am. Chem. Soc, 124: 13684-13685 (2002).

[0064] A nanoconjugate of the disclosure generally comprises a polynucleotide from about 5 nucleotides to about 100 nucleotides in length. More specifically, nanoconjugates comprise polynucleotides that are about 5 to about 90 nucleotides in length, about 5 to about 80 nucleotides in length, about 5 to about 70 nucleotides in length, about 5 to about 60 nucleotides in length, about 5 to about 50 nucleotides in length about 5 to about 45 nucleotides in length, about 5 to about 40 nucleotides in length, about 5 to about 35 nucleotides in length, about 5 to about 30 nucleotides in length, about 5 to about 25 nucleotides in length, about 5 to about 20 nucleotides in length, about 5 to about 15 nucleotides in length, about 5 to about 10 nucleotides in length, and all polynucleotides intermediate in length of the sizes specifically disclosed to the extent that the polynucleotide is able to achieve the desired result. Accordingly, polynucleotides of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or more nucleotides in length are

contemplated.

[0065] In some aspects, a polynucleotide as described herein comprises an alkyne. In various embodiments, from 1 to 100 alkyne moieties are present on a polynucleotide. In further aspects, from about 5 to about 50 alkyne moieties, or about 10 to about 20 alkyne moieties are present on a polynucleotide. In one aspect, 10 alkyne moieties are present on the polynucleotide. In further aspects, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or more alkyne moieties are present on a polynucleotide.

[0066] In another embodiment, the alkyne moieties on a polynucleotide are on the 5' end. In a further embodiment, the alkyne moieties on a polynucleotide are on the 3' end. It is contemplated that in some aspects the alkyne moieties represent only a portion of the length of a polynucleotide. By way of example, if a polynucleotide is 20 nucleotides in length, then it is contemplated that the first 10 nucleotides (counting, in various aspects from either the 5' or 3' end) comprise an alkyne moiety. Thus, 10 nucleotides comprising an alkyne moiety out of a total of 20 nucleotides results in 50% of the nucleotides in a polynucleotide being associated with an alkyne moiety. In various aspects it is contemplated that from about 0.01% to about 100% of the nucleotides in a polynucleotide are associated with an alkyne moiety. In further aspects, about 1% to about 70%, or about 2% to about 60%, or about 5% to about 50%, or about 10% to about 50%, or about 10% to about 40%, or about 20% to about 50%, or about 20% to about 40% of nucleotides in a polynucleotide are associated with an alkyne moiety.

[0067] Polynucleotides, as defined herein, also includes aptamers. The production and use of aptamers is known to those of ordinary skill in the art. In general, aptamers are nucleic acid or peptide binding species capable of tightly binding to and discreetly distinguishing target ligands [Yan et ah, RNA Biol. 6(3) 316-320 (2009), incorporated by reference herein in its entirety] . Aptamers, in some embodiments, may be obtained by a technique called the systematic evolution of ligands by exponential enrichment (SELEX) process [Tuerk et ah, Science 249:505-10 (1990), U.S. Patent Number 5,270,163, and U.S. Patent Number 5,637,459, each of which is incorporated herein by reference in their entirety]. General discussions of nucleic acid aptamers are found in, for example and without limitation, Nucleic Acid and Peptide Aptamers: Methods and Protocols (Edited by Mayer, Humana Press, 2009) and Crawford et ah, Briefings in Functional Genomics and Proteomics 2(1): 72- 79 (2003). Additional discussion of aptamers, including but not limited to selection of RNA aptamers, selection of DNA aptamers, selection of aptamers capable of covalently linking to a target protein, use of modified aptamer libraries, and the use of aptamers as a diagnostic agent and a therapeutic agent is provided in Kopylov et ah, Molecular Biology 34(6): 940- 954 (2000) translated from Molekulyarnaya Biologiya, Vol. 34, No. 6, 2000, pp. 1097-1113, which is incorporated herein by reference in its entirety. In various embodiments, an aptamer is between about 10 to about 100 nucleotides in length.

Spacers

[0068] In certain aspects, nanoconjugates are contemplated which include those wherein a nanoconjugate comprises a polynucleotide which further comprises a spacer. The spacer, in various aspects, comprises one or more crosslinking moieties as described below.

[0069] "Spacer" as used herein means a moiety that serves to contain one or more crosslinking moieties, or, in some aspects wherein the nanoconjugate comprises a

nanoparticle, increase distance between the nanoparticle and the polynucleotide, or to increase distance between individual polynucleotides when attached to the nanoparticle in multiple copies. In aspects of the disclosure wherein a nanoconjugate is used for a biological activity, it is contemplated that the spacer does not directly participate in the activity of the polynucleotide to which it is attached.

[0070] Thus, in some aspects, the spacer is contemplated herein to facilitate crosslinking via one or more crosslinking moieties. Spacers are additionally contemplated, in various aspects, as being located between individual polynucleotides in tandem, whether the polynucleotides have the same sequence or have different sequences. In one aspect, the spacer when present is an organic moiety. In another aspect, the spacer is a polymer, including but not limited to a water-soluble polymer, a nucleic acid, a polypeptide, an oligosaccharide, a carbohydrate, a lipid, or combinations thereof.

[0071] In some embodiments, the spacer is functionalized to a nanoparticle but is not linked to another biomolecule. The function of the spacer in these embodiments is to protect the nanoconjugate in vivo. Thus, in some embodiments, a spacer is functionalized to a nanoparticle. In further embodiments, the spacer is polyethylene glycol (PEG). In embodiments wherein the water soluble polymer is PEG, it is contemplated that the PEG is functionalized to a nanoparticle via a covalent bond. In some embodiments, the PEG is attached to the nanoparticle via a thiol bond. PEGylation is contemplated to protect the nanoconjugate in circulation and improve its pharmacodynamic and pharmacokinetic profiles [Harris et ah, Nat Rev Drug Discov. 2: 214-21 (2003)]. The PEGylation process attaches repeating units of ethylene glycol (polyethylene glycol (PEG)) to a nanoparticle. PEG molecules have a large hydrodynamic volume (5-10 times the size of globular proteins), are highly water soluble and hydrated, non-toxic, non-immunogenic and rapidly cleared from the body. PEGylation of nanoconjugates leads, in various embodiments, to increased resistance to enzymatic degradation, increased half-life in vivo, reduced dosing frequency, decreased immunogenicity, increased physical and thermal stability, increased solubility, increased liquid stability, and reduced aggregation.

[0072] The length of the spacer in various embodiments at least about 5 nucleotides, at least about 10 nucleotides, 10-30 nucleotides, or even greater than 30 nucleotides. The spacer may have any sequence which does not interfere with the ability of the

polynucleotides to become bound to the nanoparticles or to the target polynucleotide. The spacers should not have sequences complementary to each other or to that of the

polynucleotides, but may be all or in part complementary to the target polynucleotide. In certain aspects, the bases of the polynucleotide spacer are all adenines, all thymines, all cytidines, all guanines, all uracils, or all some other modified base.

Modified Polynucleotides

[0073] As discussed above, modified polynucleotides are contemplated for use in producing nanoconjugates. In various aspects, a polynucleotide is completely modified or partially modified. Thus, in various aspects, one or more, or all, sugar and/or one or more or all internucleotide linkages of the nucleotide units in the polynucleotide are replaced with "non-naturally occurring" groups.

[0074] In one aspect, this embodiment contemplates a peptide nucleic acid (PNA). In PNA compounds, the sugar-backbone of a polynucleotide is replaced with an amide containing backbone. See, for example US Patent Nos. 5,539,082; 5,714,331; and 5,719,262, and Nielsen et ah, Science, 1991, 254, 1497-1500, the disclosures of which are herein incorporated by reference.

[0075] Other linkages between nucleotides and unnatural nucleotides contemplated for the disclosed polynucleotides include those described in U.S. Patent Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920; U.S. Patent Publication No. 20040219565;

International Patent Publication Nos. WO 98/39352 and WO 99/14226; Mesmaeker et. al., Current Opinion in Structural Biology 5:343-355 (1995) and Susan M. Freier and Karl-Heinz Altmann, Nucleic Acids Research, 25:4429-4443 (1997), the disclosures of which are incorporated herein by reference.

[0076] Specific examples of polynucleotides include those containing modified backbones or non-natural internucleoside linkages. Polynucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. Modified polynucleotides that do not have a phosphorus atom in their internucleoside backbone are considered to be within the meaning of "polynucleotide."

[0077] Modified polynucleotide backbones containing a phosphorus atom include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates,

phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters,

selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage. Also contemplated are polynucleotides having inverted polarity comprising a single 3' to 3' linkage at the 3'-most internucleotide linkage, i.e. a single inverted nucleoside residue which may be abasic (the nucleotide is missing or has a hydroxyl group in place thereof). Salts, mixed salts and free acid forms are also contemplated.

[0078] Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, the disclosures of which are incorporated by reference herein.

[0079] Modified polynucleotide backbones that do not include a phosphorus atom have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages; siloxane backbones; sulfide, sulfoxide and sulfone backbones;

formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones;

methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts. In still other embodiments, polynucleotides are provided with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and including— CH2— NH— O— CH2— ,— CH2— N(CH3 )— O— CH2— , ,— CH2— O— N(CH3)— CH2— ,— CH2— N(CH3)—

N(CH3)— CH2— and— O— N(CH3)— CH2— CH2— described in US Patent Nos.

5,489,677, and 5,602,240. See, for example, U.S. Patent Nos. 5,034,506; 5,166,315;

5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, the disclosures of which are incorporated herein by reference in their entireties.

[0080] In various forms, the linkage between two successive monomers in the

polynucleotide consists of 2 to 4, desirably 3, groups/atoms selected from— CH2— ,— O— , — S— ,— NRH— , >C=0, >C=NRH, >C=S,— Si(R")2— ,—SO—,— S(0)2— ,— P(0)2— , — PO(BH3)— ,— P(0,S)— ,— P(S)2— ,— PO(R")— ,— PO(OCH3)— , and—

PO(NHRH)— , where RH is selected from hydrogen and Cl-4-alkyl, and R" is selected from Cl-6-alkyl and phenyl. Illustrative examples of such linkages are— CH2— CH2— CH2— ,— CH2— CO— CH2— ,— CH2— CHOH— CH2— ,— O— CH2— O— ,— O— CH2— CH2— , — O— CH2— CH=(including R5 when used as a linkage to a succeeding monomer),— CH2— CH2— O— ,— NRH— CH2— CH2— ,— CH2— CH2— NRH— ,— CH2— NRH— CH2— -,— O— CH2— CH2— NRH— ,—NRH— CO— O— ,—NRH— CO— NRH— ,— NRH— CS— NRH— ,— NRH— C(=NRH)— NRH— ,— NRH— CO— CH2— NRH— O— CO— O— ,— O— CO— CH2— O— ,— O— CH2— CO— O— ,— CH2— CO— NRH— ,— O— CO— NRH— ,— NRH— CO— CH2— ,— O— CH2— CO— NRH— ,— O— CH2— CH2— NRH— ,— CH=N— O— ,— CH2— NRH— O— ,— CH2— O— N=(including R5 when used as a linkage to a succeeding monomer),— CH2— O— NRH— ,— CO— NRH— CH2— , — CH2— NRH— O— ,— CH2— NRH— CO— ,— O— NRH— CH2— ,— O— NRH,— O— CH2— S— ,— S— CH2— O— ,— CH2— CH2— S— ,— O— CH2— CH2— S— ,— S— CH2— CH=(including R5 when used as a linkage to a succeeding monomer),— S— CH2— CH2— ,— S— CH2— CH2— O— ,— S— CH2— CH2— S— ,— CH2— S— CH2— ,— CH2— SO— CH2— ,— CH2— S02— CH2— ,— O— SO— O— ,— O— S(0)2— O— ,— O— S(0)2— CH2— ,— O— S(0)2— NRH— ,— NRH— S(0)2— CH2— ;— O— S(0)2— CH2— ,— O— P(0)2— O— ,— O— P(0,S)— O— ,— O— P(S)2— O— ,— S— P(0)2— O— , — S— P(0,S)— O— ,— S— P(S)2— O— ,— O— P(0)2— S— ,— O— P(0,S)— S— ,— O— P(S)2— S— ,— S— P(0)2— S— ,— S— P(0,S)— S— ,— S— P(S)2— S— ,— O— PO(R")— O— ,— O— PO(OCH3)— O— ,— O— PO(0 CH2CH3 )— O— ,— O— PO(0 CH2CH2S— R)— O— ,— O— PO(BH3)— O— ,— O— PO(NHRN)— O— ,— O— P(0)2— NRH H— ,— NRH— P(0)2— O— ,— O— P(0,NRH)— O— ,— CH2— P(0)2— O— ,— O— P(0)2— CH2— , and— O— Si(R")2— O— ; among which— CH2— CO— NRH— ,— CH2— NRH— O— ,— S— CH2— O— ,— O— P(0)2— O— O— P(- 0,S)— O— ,— O— P(S)2— O— ,— NRH P(0)2— O— ,— O— P(0,NRH)— O— ,— O— PO(R")— O— ,— O— PO(CH3)— O— , and— O— PO(NHRN)— O— , where RH is selected form hydrogen and Cl-4-alkyl, and R" is selected from Cl-6-alkyl and phenyl, are contemplated. Further illustrative examples are given in Mesmaeker et. al., 1995, Current Opinion in Structural Biology, 5: 343-355 and Susan M. Freier and Karl-Heinz Altmann, 1997, Nucleic Acids Research, vol 25: pp 4429- 4443.

[0081] Still other modified forms of polynucleotides are described in detail in U.S. Patent Application No. 20040219565, the disclosure of which is incorporated by reference herein in its entirety.

[0082] Modified polynucleotides may also contain one or more substituted sugar moieties. In certain aspects, polynucleotides comprise one of the following at the 2' position: OH; F; O- , S-, or N-alkyl; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted CI to CIO alkyl or C2 to CIO alkenyl and alkynyl. Other embodiments include 0[(CH2)nO]mCH3, 0(CH2)nOCH3, 0(CH2)nNH2, 0(CH2)nCH3, 0(CH2)nONH2, and 0(CH2)nON[(CH2)nCH3]2, where n and m are from 1 to about 10. Other polynucleotides comprise one of the following at the 2' position: CI to CIO lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O- alkaryl or O-aralkyl, SH, SCH3, OCN, CI, Br, CN, CF3, OCF3, SOCH3, S02CH3, ON02, N02, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of a polynucleotide, or a group for improving the pharmacodynamic properties of a polynucleotide, and other substituents having similar properties. In one aspect, a modification includes 2'-methoxyethoxy (2'-0-CH2CH20CH3, also known as 2'-0-(2-methoxyethyl) or 2'-MOE) (Martin et al, 1995, Helv. Chim. Acta, 78: 486-504) i.e., an alkoxyalkoxy group. Other modifications include 2'- dimethylaminooxyethoxy, i.e., a 0(CH2)20N(CH3)2 group, also known as 2'-DMAOE, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-0-dimethyl-amino-ethoxy-ethyl or 2'-DMAEOE), i.e., 2'-0— CH2— O— CH2— N(CH3)2.

[0083] Still other modifications include 2'-methoxy (2'-0— CH3), 2'-aminopropoxy (2'- OCH2CH2CH2NH2), 2'-allyl (2'-CH2— CH=CH2), 2'-0-allyl (2'-0— CH2— CH=CH2) and 2'-fluoro (2'-F). The 2'-modification may be in the arabino (up) position or ribo (down) position. In one aspect, a 2'-arabino modification is 2'-F. Similar modifications may also be made at other positions on the polynucleotide, for example, at the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked polynucleotides and the 5' position of 5' terminal nucleotide. Polynucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. See, for example, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920, the disclosures of which are incorporated by reference in their entireties herein.

[0084] In one aspect, a modification of the sugar includes Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3' or 4' carbon atom of the sugar ring, thereby forming a bicyclic sugar moiety. The linkage is in certain aspects a methylene (— CH2— ) n group bridging the 2' oxygen atom and the 4' carbon atom wherein n is 1 or 2. LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226, the disclosures of which are incorporated herein by reference.

Polynucleotide Features

[0085] A nanoconjugate of the disclosure, in various aspects, comprises a plurality of polynucleotides. As a result, each nanoconjugate has the ability to bind to a plurality of target polynucleotides having a sufficiently complementary sequence. For example, if a specific polynucleotide is targeted, a single nanoconjugate has the ability to bind to multiple copies of the same molecule. In one aspect, methods are provided wherein the nanoconjugate comprises identical polynucleotides, i.e., each polynucleotide has the same length and the same sequence. In other aspects, the nanoconjugate comprises two or more polynucleotides which are not identical, i.e., at least one of the polynucleotides of the nanoconjugate differ from at least one other polynucleotide of the nanoconjugate in that it has a different length and/or a different sequence. In aspects wherein a nanoconjugate comprises different polynucleotides, these different polynucleotides bind to the same single target polynucleotide but at different locations, or bind to different target polynucleotides which encode different gene products. Accordingly, in various aspects, a single nanoconjugate may be used in a method to inhibit expression of more than one gene product. Polynucleotides are thus used to target specific polynucleotides, whether at one or more specific regions in the target polynucleotide, or over the entire length of the target polynucleotide as the need may be to effect a desired level of inhibition of gene expression.

[0086] Accordingly, in one aspect, the polynucleotides are designed with knowledge of the target sequence. Alternatively, a polynucleotide in a nanoconjugate need not hybridize to a target polynucleotide in order to achieve a desired effect as described herein.

[0087] Polynucleotides contemplated for production of a nanoconjugate include, in one aspect, those which modulate expression of a gene product expressed from a target polynucleotide. Accordingly, antisense polynucleotides which hybridize to a target polynucleotide and inhibit translation, siRNA polynucleotides which hybridize to a target polynucleotide and initiate an RNAse activity (for example RNAse H), triple helix forming polynucleotides which hybridize to double- stranded polynucleotides and inhibit transcription, and ribozymes which hybridize to a target polynucleotide and inhibit translation, are contemplated.

[0088] In some aspects, a polynucleotide-based nanoconjugate allows for efficient uptake of the nanoconjugate. In various aspects, the polynucleotide comprises a nucleotide sequence that allows increased uptake efficiency of the nanoconjugate. As used herein, "efficiency" refers to the number or rate of uptake of nanoconjugates in/by a cell. Because the process of nanoconjugates entering and exiting a cell is a dynamic one, efficiency can be increased by taking up more nanoconjugates or by retaining those nanoconjugates that enter the cell for a longer period of time. Similarly, efficiency can be decreased by taking up fewer

nanoconjugates or by retaining those nanoconjugates that enter the cell for a shorter period of time.

[0089] Thus, the nucleotide sequence can be any nucleotide sequence that is desired may be selected for, in various aspects, increasing or decreasing cellular uptake of a

nanoconjugate or gene regulation. The nucleotide sequence, in some aspects, comprises a homopolymeric sequence which affects the efficiency with which the nanoparticle to which the polynucleotide is attached is taken up by a cell. Accordingly, the homopolymeric sequence increases or decreases the efficiency. It is also contemplated that, in various aspects, the nucleotide sequence is a combination of nucleobases, such that it is not strictly a homopolymeric sequence. For example and without limitation, in various aspects, the nucleotide sequence comprises alternating thymidine and uridine residues, two thymidines followed by two uridines or any combination that affects increased uptake is contemplated by the disclosure. In some aspects, the nucleotide sequence affecting uptake efficiency is included as a domain in a polynucleotide comprising additional sequence. This "domain" would serve to function as the feature affecting uptake efficiency, while the additional nucleotide sequence would serve to function, for example and without limitation, to regulate gene expression. In various aspects, the domain in the polynucleotide can be in either a proximal, distal, or center location relative to the nanoconjugate. It is also contemplated that a polynucleotide comprises more than one domain.

[0090] The homopolymeric sequence, in some embodiments, increases the efficiency of uptake of the nanoconjugate by a cell. In some aspects, the homopolymeric sequence comprises a sequence of thymidine residues (polyT) or uridine residues (polyU). In further aspects, the polyT or polyU sequence comprises two thymidines or uridines. In various aspects, the polyT or polyU sequence comprises from about 3 to about 500 thymidine or uridine residues. In further embodiments, the polyT or polyU sequence comprises from about 3 to about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, about 100, about 150, about 200 or more thymidine or uridine residues. In some embodiments, the polyT or polyU sequences comprises from about 10 to about 50, about 20 to about 100, or about 40 to about 200 thymidine or uridine residues. Accordingly, in various embodiments, the polyT or polyU sequence comprises 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 125, about 150, about 175, about 200, about 250, about 300, about 350, about 400, about 450, about 500 or more thymidine or uridine residues.

[0091] In some embodiments, it is contemplated that a nanoconjugate comprising a polynucleotide that comprises a homopolymeric sequence is taken up by a cell with greater efficiency than a nanoconjugate comprising the same polynucleotide but lacking the homopolymeric sequence. In various aspects, a nanoconjugate comprising a polynucleotide that comprises a homopolymeric sequence is taken up by a cell about 2-fold, about 3-fold, about 4-fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10- fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold or higher, more efficiently than a nanoconjugate comprising the same polynucleotide but lacking the homopolymeric sequence.

[0092] In other aspects, the domain is a phosphate polymer (C3 residue). In some aspects, the domain comprises a phosphate polymer (C3 residue) that is comprised of two phosphates. In some embodiments, the C3 residue comprises from about 3 to about 500, or from about 5 to about 50 phosphates, or from about 10 to about 50 phosphates, or from about 20 to about 70 phosphates, or from about 50 to about 200 phosphates. In various aspects, the C3 residue comprises 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 125, about 150, about 175, about 200, about 250, about 300, about 350, about 400, about 450, about 500 or more phosphates.

[0093] In some embodiments, it is contemplated that a nanoconjugate comprising a polynucleotide which comprises a domain is taken up by a cell with lower efficiency than a nanoconjugate comprising the same polynucleotide but lacking the domain. In various aspects, a nanoconjugate comprising a polynucleotide which comprises a domain is taken up by a cell about 2-fold, about 3-fold, about 4-fold, about 5-fold, about 6-fold, about 7-fold, about 8-fold, about 9-fold, about 10-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold or higher, less efficiently than a nanoconjugate comprising the same polynucleotide but lacking the domain.

[0094] A surface density adequate to make the nanoconjugates stable and the conditions necessary to obtain it for a desired combination of nanoconjugates and polynucleotides can be determined empirically. Generally, a surface density of at least 2 pmol/cm will be adequate to provide stable nanoconjugate-polynucleotide compositions. In some aspects, the surface density is at least 15 pmol/cm . In additional aspects, the polynucleotide is associated with the nanoconjugate at a surface density of about 0.3 pmol/cm 2 to about 10 pmol/cm 2 , or from about 0.6 pmol/cm 2 to about 15 pmol/cm 2 , or from about 1 pmol/cm 2 to about 20 pmol/cm 2 , or from about 0.3 pmol/cm 2 to about 100 pmol/cm 2. Methods are also provided wherein the polynucleotide is associate with the nanoconjugate at a surface density of at least

2 pmol/cm 2 , at least 3 pmol/cm 2 , at least 4 pmol/cm 2 , at least 5 pmol/cm 2 , at least 6 pmol/cm 2 , at least 7 pmol/cm 2 , at least 8 pmol/cm 2 , at least 9 pmol/cm 2 , at least 10 pmol/cm 2 , at least about 15 pmol/cm 2 , at least about 19 pmol/cm 2 , at least about 20 pmol/cm 2 , at least about 25 pmol/cm 2 , at least about 30 pmol/cm 2 , at least about 35 pmol/cm 2 , at least about 40 pmol/cm 2 , at least about 45 pmol/cm 2 , at least about 50 pmol/cm 2 , at least about 55 pmol/cm 2 , at least about 60 pmol/cm 2 , at least about 65 pmol/cm 2 , at least about 70 pmol/cm 2 , at least about 75 pmol/cm 2 , at least about 80 pmol/cm 2 , at least about 85 pmol/cm 2 , at least about 90 pmol/cm 2 , at least about 95 pmol/cm 2 , at least about 100 pmol/cm 2 , at least about 125 pmol/cm 2 , at least about 150 pmol/cm 2 , at least about 175 pmol/cm 2 , at least about 200 pmol/cm 2 , at least about 250 pmol/cm 2 , at least about 300 pmol/cm 2 , at least about 350 pmol/cm 2 , at least about 400 pmol/cm 2 , at least about 450 pmol/cm 2 , at least about 500 pmol/cm 2 , at least about 550 pmol/cm 2 , at least about 600 pmol/cm 2 , at least about 650 pmol/cm 2 , at least about 700 pmol/cm 2 , at least about 750 pmol/cm 2 , at least about 800 pmol/cm 2 , at least about 850 pmol/cm 2 , at least about 900 pmol/cm 2 , at least about 950 pmol/cm 2 , at least about 1000 pmol/cm 2 or more.

[0095] As used herein, a "conjugation site" is understood to mean a site on a

polynucleotide to which a contrast agent is attached. Methods of attaching a contrast agent to a polynucleotide are generally known in the art [see, for example, Song et ah, Chem Ing Engl 48(48): 9143-9147 (2009)]. In certain aspects, the disclosure also provides one or more polynucleotides that are part of the nanoconjugate do not comprise a conjugation site while one or more polynucleotides that are part of the same nanoconjugate do comprise a conjugation site. Conjugation of a contrast agent to a nanoconjugate through a conjugation site is generally described in PCT/US2010/44844, which is incorporated herein by reference in its entirety. The disclosure provides, in one aspect, a nanoconjugate comprising a polynucleotide wherein the polynucleotide comprises one to about ten conjugation sites. In another aspect, the polynucleotide comprises five conjugation sites. In general, for a nucleotide, both its backbone (phosphate group) and nucleobase can be modified.

Accordingly, the present disclosure contemplates that there are 2n conjugation sites, where n=length of the polynucleotide template. In related aspects, it is contemplated that the composition comprises a nanoconjugate comprising a plurality of polynucleotides. In some aspects, the plurality of polynucleotides comprises at least one polynucleotide to which contrast agents are associated through one or more conjugation sites, as well as at least one polynucleotide that has gene regulatory activity as described herein. [0096] Accordingly, in some embodiments, it is contemplated that one or more polynucleotides that are part of the nanoconjugate is not conjugated to a contrast agent while one or more polynucleotides that are part of the same nanoconjugate are conjugated to a contrast agent.

Polynucleotide marker/label

[0097] A polynucleotide as described herein, in various aspects, optionally comprises a detectable label. Accordingly, the disclosure provides compositions and methods wherein polynucleotide hybridization is detected by a detectable change. In one aspect, hybridization gives rise to a color change which is observed with the naked eye or spectroscopically.

[0098] Methods for visualizing the detectable change resulting from polynucleotide hybridization also include any fluorescent detection method, including without limitation fluorescence microscopy, a microtiter plate reader or fluorescence-activated cell sorting (FACS).

[0099] It will be understood that a label contemplated by the disclosure includes any of the fluorophores described herein as well as other detectable labels known in the art. For example, labels also include, but are not limited to, redox active probes, chemiluminescent molecules, radioactive labels, dyes, fluorescent molecules, phosphorescent molecules, imaging and/or contrast agents as described below, quantum dots, as well as any marker which can be detected using spectroscopic means, i.e., those markers detectable using microscopy and cytometry. In aspects of the disclosure wherein a detectable label is to be detected, the disclosure provides that any luminescent, fluorescent, or phosphorescent molecule or particle can be efficiently quenched by noble metal surfaces, or by a quencher molecule known in the art (quencher molecules contemplated by the disclosure include but are not limited to Dabsyl (dimethylaminoazobenzenesulfonic acid), Black Hole Quenchers, Qxl quenchers, Iowa black FQ, Iowa black RQ, IRDye QC-1 and a combination thereof). Accordingly, each type of molecule is contemplated for use in the compositions and methods disclosed.

[0100] Methods of labeling biomolecules with fluorescent molecules and measuring fluorescence are well known in the art.

[0101] Suitable fluorescent molecules are also well known in the art and include without limitation 1,8-ANS (l-Anilinonaphthalene-8-sulfonic acid), l-Anilinonaphthalene-8-sulfonic acid (1,8-ANS), 5-(and-6)-Carboxy-2', V-dichlorofluorescein pH 9.0, 5-FAM pH 9.0, 5-ROX (5-Carboxy-X-rhodamine, triethylammonium salt), 5-ROX pH 7.0, 5-TAMRA, 5 -T AMR A pH 7.0, 5-TAMRA-MeOH, 6 JOE, 6,8-Difluoro-7-hydroxy-4-methylcoumarin pH 9.0, 6-

Carboxyrhodamine 6G pH 7.0, 6-Carboxyrhodamine 6G, hydrochloride, 6-HEX, SE pH 9.0,

6-TET, SE pH 9.0, 7-Amino-4-methylcoumarin pH 7.0, 7-Hydroxy-4-methylcoumarin, 7-

Hydroxy-4-methylcoumarin pH 9.0, Alexa 350, Alexa 405, Alexa 430, Alexa 488, Alexa

532, Alexa 546, Alexa 555, Alexa 568, Alexa 594, Alexa 647, Alexa 660, Alexa 680, Alexa

700, Alexa Fluor 430 antibody conjugate pH 7.2, Alexa Fluor 488 antibody conjugate pH 8.0,

Alexa Fluor 488 hydrazide-water, Alexa Fluor 532 antibody conjugate pH 7.2, Alexa Fluor

555 antibody conjugate pH 7.2, Alexa Fluor 568 antibody conjugate pH 7.2, Alexa Fluor 610

R-phycoerythrin streptavidin pH 7.2, Alexa Fluor 647 antibody conjugate pH 7.2, Alexa

Fluor 647 R-phycoerythrin streptavidin pH 7.2, Alexa Fluor 660 antibody conjugate pH 7.2,

Alexa Fluor 680 antibody conjugate pH 7.2, Alexa Fluor 700 antibody conjugate pH 7.2,

Allophycocyanin pH 7.5, AMCA conjugate, Amino Coumarin, APC (allophycocyanin) ,Atto

647, BCECF pH 5.5, BCECF pH 9.0, BFP (Blue Fluorescent Protein), BO-PRO- 1-DNA,

BO-PRO-3-DNA, BOBO-l-DNA, BOBO-3-DNA, BODJPY 650/665-X, MeOH, BODIPY

FL conjugate, BODIPY FL, MeOH, Bodipy R6G SE, BODIPY R6G, MeOH, BODIPY

TMR-X antibody conjugate pH 7.2, Bodipy TMR-X conjugate, BODIPY TMR-X, MeOH,

BODIPY TMR-X, SE, BODIPY TR-X phallacidin pH 7.0, BODIPY TR-X, MeOH,

BODIPY TR-X, SE, BOPRO-1, BOPRO-3, Calcein, Calcein pH 9.0, Calcium Crimson,

Calcium Crimson Ca2+, Calcium Green, Calcium Green- 1 Ca2+, Calcium Orange, Calcium

Orange Ca2+, Carboxynaphthofluorescein pH 10.0, Cascade Blue, Cascade Blue BSA pH

7.0, Cascade Yellow, Cascade Yellow antibody conjugate pH 8.0, CFDA, CFP (Cyan

Fluorescent Protein), CI-NERF pH 2.5, CI-NERF pH 6.0, Citrine, Coumarin, Cy 2, Cy 3, Cy

3.5, Cy 5, Cy 5.5, CyQUANT GR-DNA, Dansyl Cadaverine, Dansyl Cadaverine, MeOH,

DAPI, DAPI-DNA, Dapoxyl (2-aminoethyl) sulfonamide, DDAO pH 9.0, Di-8 ANEPPS, Di-

8-ANEPPS-lipid, Dil, DiO, DM-NERF pH 4.0, DM-NERF pH 7.0, DsRed, DTAF, dTomato, eCFP (Enhanced Cyan Fluorescent Protein), eGFP (Enhanced Green Fluorescent Protein),

Eosin, Eosin antibody conjugate pH 8.0, Erythrosin-5-isothiocyanate pH 9.0, Ethidium

Bromide, Ethidium homodimer, Ethidium homodimer- 1-DNA, eYFP (Enhanced Yellow

Fluorescent Protein), FDA, FITC, FITC antibody conjugate pH 8.0, FlAsH, Fluo-3, Fluo-3

Ca2+, Fluo-4, Fluor-Ruby, Fluorescein, Fluorescein 0.1 M NaOH, Fluorescein antibody conjugate pH 8.0, Fluorescein dextran pH 8.0, Fluorescein pH 9.0, Fluoro-Emerald, FM 1-43,

FM 1-43 lipid, FM 4-64, FM 4-64, 2% CHAPS, Fura Red Ca2+, Fura Red, high Ca, Fura

Red, low Ca, Fura-2 Ca2+, Fura-2, high Ca, Fura-2, no Ca, GFP (S65T), HcRed, Hoechst 33258, Hoechst 33258-DNA, Hoechst 33342, Indo-1 Ca2+, Indo-1, Ca free, Indo-1, Ca saturated, JC-1, JC-1 pH 8.2, Lissamine rhodamine, LOLO-l-DNA, Lucifer Yellow, CH, LysoSensor Blue, LysoSensor Blue pH 5.0, LysoSensor Green, LysoSensor Green pH 5.0, LysoSensor Yellow pH 3.0, LysoSensor Yellow pH 9.0, LysoTracker Blue, LysoTracker Green, LysoTracker Red, Magnesium Green, Magnesium Green Mg2+, Magnesium Orange, Marina Blue, mBanana, mCherry, mHoneydew, MitoTracker Green, MitoTracker Green FM, MeOH, MitoTracker Orange, MitoTracker Orange, MeOH, MitoTracker Red, MitoTracker Red, MeOH, mOrange, mPlum, mRFP, mStrawberry, mTangerine, NBD-X, NBD-X, MeOH, NeuroTrace 500/525, green fluorescent Nissl stain-RNA, Nile Blue, EtOH, Nile Red, Nile Red-lipid, Nissl, Oregon Green 488, Oregon Green 488 antibody conjugate pH 8.0, Oregon Green 514, Oregon Green 514 antibody conjugate pH 8.0, Pacific Blue, Pacific Blue antibody conjugate pH 8.0, Phycoerythrin, PicoGreen dsDNA quantitation reagent, PO-PRO-1, PO- PRO-l-DNA, PO-PRO-3, PO-PRO-3-DNA, POPO-1, POPO-l-DNA, POPO-3, Propidium Iodide, Propidium Iodide-DNA, R-Phycoerythrin pH 7.5, ReAsH, Resorufin, Resorufin pH 9.0, Rhod-2, Rhod-2 Ca2+, Rhodamine, Rhodamine 110, Rhodamine 110 pH 7.0, Rhodamine 123, MeOH, Rhodamine Green, Rhodamine phalloidin pH 7.0, Rhodamine Red-X antibody conjugate pH 8.0, Rhodaminen Green pH 7.0, Rhodol Green antibody conjugate pH 8.0, Sapphire, SBFI-Na+, Sodium Green Na+, Sulforhodamine 101, EtOH, SYBR Green I, SYPRO Ruby, SYTO 13-DNA, SYTO 45-DNA, SYTOX Blue-DNA, Tetramethylrhodamine antibody conjugate pH 8.0, Tetramethylrhodamine dextran pH 7.0, Texas Red-X antibody conjugate pH 7.2, TO-PRO-l-DNA, TO-PRO-3-DNA, TOTO-l-DNA, TOTO-3-DNA, TRITC, X-Rhod-1 Ca2+, YO-PRO-l-DNA, YO-PRO-3-DNA, YOYO-l-DNA, and YOYO- 3-DNA.

Polypeptides

[0102] Nanoconjugates, in various aspects, comprise a polypeptide. The polypeptide may be associated with the nanoconjugate or may be delivered in a composition with a

nanoconjugate as, in some embodiments, a therapeutic agent. As used herein a "polypeptide" refers to a polymer comprised of amino acid residues. Polypeptides are understood in the art and include without limitation an antibody, an enzyme and a hormone. In related aspects, the nanoconjugate comprising a polypeptide recognizes and associates with a target molecule and enables detection of the target molecule.

[0103] Polypeptides of the disclosure may be either naturally occurring or non-naturally occurring. Polypeptides optionally include a spacer as described herein above. Naturally Occurring Polypeptides

[0104] Naturally occurring polypeptides include without limitation biologically active polypeptides (including antibodies) that exist in nature or can be produced in a form that is found in nature by, for example, chemical synthesis or recombinant expression techniques. Naturally occurring polypeptides also include lipoproteins and post-translationally modified proteins, such as, for example and without limitation, glycosylated proteins.

[0105] Antibodies contemplated for use in the methods and compositions of the present disclosure include without limitation antibodies that recognize and associate with a target molecule either in vivo or in vitro.

Non-Naturally Occurring Polypeptides

[0106] Non-naturally occurring polypeptides contemplated by the present disclosure include but are not limited to synthetic polypeptides, as well as fragments, analogs and variants of naturally occurring or non-naturally occurring polypeptides as defined herein. Non-naturally occurring polypeptides also include proteins or protein substances that have D- amino acids, modified, derivatized, or non-naturally occurring amino acids in the D- or reconfiguration and/or peptidomimetic units as part of their structure. The term "protein" typically refers to large polypeptides. The term "peptide" typically refers to short (i.e., equal to or less than about 50 amino acids) polypeptides.

[0107] Non-naturally occurring polypeptides are prepared, for example, using an automated polypeptide synthesizer or, alternatively, using recombinant expression techniques using a modified polynucleotide which encodes the desired polypeptide.

[0108] As used herein a "fragment" of a polypeptide is meant to refer to any portion of a polypeptide or protein smaller than the full-length polypeptide or protein expression product.

[0109] As used herein an "analog" refers to any of two or more polypeptides substantially similar in structure and having the same biological activity, but can have varying degrees of activity, to either the entire molecule, or to a fragment thereof. Analogs differ in the composition of their amino acid sequences based on one or more mutations involving substitution, deletion, insertion and/or addition of one or more amino acids for other amino acids. Substitutions can be conservative or non-conservative based on the physico-chemical or functional relatedness of the amino acid that is being replaced and the amino acid replacing it. [0110] As used herein a "variant" refers to a polypeptide, protein or analog thereof that is modified to comprise additional chemical moieties not normally a part of the molecule. Such moieties may modulate, for example and without limitation, the molecule's solubility, absorption, and/or biological half-life. Moieties capable of mediating such effects are disclosed in Remington's Pharmaceutical Sciences (1980). Procedures for coupling such moieties to a molecule are well known in the art. In various aspects, polypeptides are modified by glycosylation, PEGylation, and/or polysialylation.

[0111] Fusion proteins, including fusion proteins wherein one fusion component is a fragment or a mimetic, are also contemplated. A "mimetic" as used herein means a peptide or protein having a biological activity that is comparable to the protein of which it is a mimetic. By way of example, an endothelial growth factor mimetic is a peptide or protein that has a biological activity comparable to the native endothelial growth factor. The term further includes peptides or proteins that indirectly mimic the activity of a protein of interest, such as by potentiating the effects of the natural ligand of the protein of interest.

[0112] Polypeptides include antibodies along with fragments and derivatives thereof, including but not limited to Fab' fragments, F(ab)2 fragments, Fv fragments, Fc fragments , one or more complementarity determining regions (CDR) fragments, individual heavy chains, individual light chain, dimeric heavy and light chains (as opposed to heterotetrameric heavy and light chains found in an intact antibody, single chain antibodies (scAb), humanized antibodies (as well as antibodies modified in the manner of humanized antibodies but with the resulting antibody more closely resembling an antibody in a non-human species), chelating recombinant antibodies (CRABs), bispecific antibodies and multispecific antibodies, and other antibody derivative or fragments known in the art.

Contrast agents

[0113] Disclosed herein are, in various aspects, methods and compositions comprising a nanoconjugate, wherein a biomolecule is conjugated to a contrast agent through a conjugation site. In various aspects, a contrast agent is conjugated to a polynucleotide and/or a polypeptide. As used herein, a "contrast agent" is a compound or other substance introduced into a cell in order to create a difference in the apparent density of various organs and tissues, making it easier to see the delineate adjacent body tissues and organs. It will be understood that conjugation of a contrast agent to a polynucleotide or polypeptide described herein is useful in the compositions and methods of the disclosure. [0114] Methods provided by the disclosure include those wherein relaxivity of the contrast agent in association with a nanoconjugate is increased relative to the relaxivity of the contrast agent in the absence of being associated with a nanoconjugate. In some aspects, the increase is about 1- fold to about 20-fold. In further aspects, the increase is about 2-fold fold to about 10-fold, and in yet further aspects the increase is about 3-fold.

[0115] In some embodiments, the contrast agent is selected from the group consisting of gadolinium, xenon, iron oxide, a manganese chelate (Mn-DPDP) and copper. Thus, in some embodiments the contrast agent is a paramagnetic compound, and in some aspects, the paramagnetic compound is gadolinium.

[0116] The present disclosure also contemplates contrast agents that are useful for positron emission tomography (PET) scanning. In some aspects, the PET contrast agent is a radionuclide. In certain embodiments the contrast agent comprises a PET contrast agent comprising a label selected from the group consisting of n C, 13 N, 18 F, ^Cu, 68 Ge, 99m Tc and

82 Ru. In particular embodiments the contrast agent is a PET contrast agent selected from the group consisting of [ u C]choline, [ 18 F]fluorodeoxyglucose(FDG), [ x ^methionine,

[ n C]choline, [ n C]acetate, [ 18 F]fluorocholine, ^Cu chelates, 99m Tc chelates, and

[ 18 F]polyethyleneglycol stilbenes.

[0117] The disclosure also provides methods wherein a PET contrast agent is introduced into a polynucleotide during the polynucleotide synthesis process or is conjugated to a nucleotide following polynucleotide synthesis. For example and without limitation, nucleotides can be synthesized in which one of the phosphorus atoms is replaced with 32 P or

33 P, one of the oxygen atoms in the phosphate group is replaced with 35 S, or one or more of the hydrogen atoms is replaced with H. A functional group containing a radionuclide can also be conjugated to a nucleotide through conjugation sites.

[0118] In certain embodiments, the MRI contrast agent conjugated to a polynucleotide is iron or paramagnetic radiotracers and/or complexes, including but not limited to gadolinium, xenon, iron oxide, and copper. MRI contrast agents can include, but are not limited to positive contrast agents and/or negative contrast agents. Positive contrast agents cause a reduction in the T \ relaxation time (increased signal intensity on T \ weighted images). They (appearing bright on MRI) are typically small molecular weight compounds containing as their active element Gadolinium, Manganese, or Iron. All of these elements have unpaired electron spins in their outer shells and long relaxivities. A special group of negative contrast agents (appearing dark on MRI) include perfluorocarbons (perfluorochemicals), because their presence excludes the hydrogen atoms responsible for the signal in MR imaging.

[0119] The composition of the disclosure, in various aspects, is contemplated to comprise a nanoconjugate that comprises about 50 to about 2.5 X 10 6 contrast agents. In some embodiments, the nanoconjugate comprises about 500 to about 1 X 10 6 contrast agents.

Targeting Moiety

[0120] The term "targeting moiety" as used herein refers to any molecular structure which assists a compound or other molecule in binding or otherwise localizing to a particular target, a target area, entering target cell(s), or binding to a target receptor. For example and without limitation, targeting moieties may include proteins, including antibodies and protein fragments capable of binding to a desired target site in vivo or in vitro, peptides, small molecules, anticancer agents, polynucleotide-binding agents, carbohydrates, ligands for cell surface receptors, aptamers, lipids (including cationic, neutral, and steroidal lipids, virosomes, and liposomes), antibodies and hormones may serve as targeting moieties.

Targeting moieties are useful for delivery of the nanoconjugate to specific cell types, as well as sub-cellular locations.

[0121] Receptor-mediated transport mechanisms are present at the BBB, and these involve the vesicular trafficking system of the brain endothelium [Jones et al., Pharm Res. 24(9): 1759-1771 (2007)]. Brain influx of nutrients such as iron [Jefferies et al., Nature 312: 162- 163 (1984)], insulin [Duffy et al., Brain Res 420: 32-38 (1987)], and leptin [Golden et al., J Clin Invest 99: 14-18 (1997)] occurs by a transcellular, receptor-mediated transport mechanism known as transcytosis.

[0122] In some embodiments, the targeting moiety is a protein. The protein portion of the composition of the present disclosure is, in some aspects, a protein capable of targeting the composition to a target cell. The targeting protein of the present disclosure may bind to a receptor, substrate, antigenic determinant, or other binding site on a target cell or other target site.

[0123] Antibodies useful as targeting proteins may be polyclonal or monoclonal. A number of monoclonal antibodies (MAbs) that bind to a specific type of cell have been developed. Antibodies derived through genetic engineering or protein engineering may be used as well (e.g., IgG, IgA, IgM, IgD, IgE antibodies). [0124] The antibody employed as a targeting agent in the present disclosure may be an intact molecule, a fragment thereof, or a functional equivalent thereof. Examples of antibody fragments useful in the compositions of the present disclosure are F(ab') 2 , Fab' Fab and Fv fragments, which may be produced by conventional methods or by genetic or protein engineering.

[0125] In additional aspects, targeting moieties contemplated by the disclosure include, but are not limited to sugars (e.g., mannose, mannose-6-phosphate, galactose). In further aspects, the moiety targets any one or a combination of the transferrin receptor (TfR), which is highly expressed by brain capillaries to mediate the delivery of iron to the brain [Jefferies et al., Nature 312: 162-163 (1984)]; the insulin receptor and insulin-like growth factor receptor [Duffy et al., Brain Res 420:32-38 (1987)]; the low density lipoprotein receptor-related protein 1 and low density lipoprotein receptor-related protein 2 [Gaillard et al., Expert Opin Drug Deliv 2:299-309 (2005)]; and the diphtheria toxin receptor/heparin binding epidermal growth factor-like growth factor [Gaillard et al., Int Congres Series 1277: 185-198 (2005)]. Additional moieties contemplated by the disclosure that are capable of effecting receptor- mediated transcytosis (RMT) include, but are not limited to, those disclosed in Feng et al. [In: Drug Delivery to the Central Nervous System, Kewal K. Jain (Editor), vol. 45: 15-34 (2010)].

[0126] In some embodiments, the polynucleotide portion of the nanoconjugate may serve as an additional or auxiliary targeting moiety. The polynucleotide portion may be selected or designed to assist in extracellular targeting, or to act as an intracellular targeting moiety. That is, the polynucleotide portion may act as a DNA probe seeking out target cells. This additional targeting capability will serve to improve specificity in delivery of the composition to target cells. The polynucleotide may additionally or alternatively be selected or designed to target the composition within target cells, while the targeting protein targets the conjugate extracellularly.

[0127] It is contemplated that the targeting moiety can, in various embodiments, be associated with a nanoconjugate. In aspects wherein the nanoconjugate comprises a nanoparticle (i.e., is not hollow), it is contemplated that the targeting moiety is attached to either the nanoparticle, the polynucleotide/polypeptide or both. In further aspects, the targeting moiety is associated with the nanoconjugate composition, and in other aspects the targeting moiety is administered before, concurrent with, or after the administration of a composition of the disclosure. Therapeutic agents

[0128] In any of the aspects or embodiments of the disclosure, it is contemplated that a therapeutic agent is delivered with a nanoconjugate. Such delivery can be, in various embodiments, facilitated by associating the therapeutic agent with a nanoconjugate, or delivery can be facilitated by co-administering the therapeutic agent with a nanoconjugate. Methods for associating a therapeutic agent to a nanoconjugate are known in the art and described, for example and without limitation, in International Patent Application Number PCT/US2010/055018, which is incorporated by reference herein in its entirety. In some embodiments, the therapeutic agent is a neurotrophic factor.

Neurotrophins

[0129] Many neurotrophic factors are neuroprotective in brain, but do not cross the blood- brain barrier. These factors are suitable for use in the compositions and methods of the disclosure and include brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-4/5, fibroblast growth factor (FGF)-2 and other FGFs, neurotrophin (NT)-3, erythropoietin (EPO), hepatocyte growth factor (HGF), epidermal growth factor (EGF), transforming growth factor (TGF)-a, TGF-β, vascular endothelial growth factor (VEGF), interleukin-1 receptor antagonist (IL- lra), ciliary neurotrophic factor (CNTF), glial- derived neurotrophic factor (GDNF), neurturin, platelet-derived growth factor (PDGF), heregulin, neuregulin, artemin, persephin, interleukins, granulocyte-colony stimulating factor (CSF), granulocyte-macrophage-CSF, netrins, cardiotrophin- 1, hedgehogs, leukemia inhibitory factor (LIF), midkine, pleiotrophin, bone morphogenetic proteins (BMPs), netrins, saposins, semaphorins, and stem cell factor (SCF).

Anticancer agent

[0130] In some aspects, a composition of the disclosure comprises an anticancer agent. Suitable anticancer agents include, but are not limited to, Actinomycin D, Alemtuzumab, Allopurinol sodium, Amifostine, Amsacrine, Anastrozole, Ara-CMP, Asparaginase,

Azacytadine, Bendamustine, Bevacizumab, Bicalutimide, Bleomycin (e.g. , Bleomycin A 2 and B 2 ), Bortezomib, Busulfan, Camptothecin sodium salt, Capecitabine, Carboplatin, Carmustine, Cetuximab, Chlorambucil, Cisplatin, Cladribine, Clofarabine,

Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Daunorubicin, Daunorubicin liposomal, Dacarbazine, Decitabine, Docetaxel, Doxorubicin, Doxorubicin liposomal, Epirubicin, Estramustine, Etoposide, Etoposide phosphate, Exemestane, Floxuridine, Fludarabine, Fluadarabine phosphate, 5-Fluorouracil, Fotemustine, Fulvestrant, Gemcitabine, Goserelin, Hexamethylmelamine, Hydroxyurea, Idarubicin, Ifosfamide, Imatinib, Irinotecan, Ixabepilone, Lapatinib, Letrozole, Leuprolide acetate, Lomustine, Mechlorethamine,

Melphalan, 6-Mercaptopurine, Methotrexate, Mithramycin, Mitomycin C, Mitotane,

Mitoxantrone, Nimustine, Ofatumumab, Oxaliplatin, Paclitaxel, Panitumumab, Pegaspargase, Pemetrexed, Pentostatin, Pertuzumab, Picoplatin, Pipobroman, Plerixafor, Procarbazine, Raltitrexed, Rituximab, Streptozocin, Temozolomide, Teniposide, 6-Thioguanine, Thiotepa, Topotecan, Trastuzumab, Treosulfan, Triethylenemelamine, Trimetrexate, Uracil Nitrogen Mustard, Valrubicin, Vinblastine, Vincristine, Vindesine, Vinorelbine, and analogues, precursors, derivatives and pro-drugs thereof. It is noted that two or more of the above compounds may be used in combination in the compositions of the disclosure.

Small molecule

[0131] The term "small molecule," as used herein, refers to a chemical compound, for instance a peptidometic that may optionally be derivatized, or any other low molecular weight organic compound, either natural or synthetic. Such small molecules may be a therapeutically deliverable substance or may be further derivatized to facilitate delivery.

[0132] By "low molecular weight" is meant compounds having a molecular weight of less than 1000 Daltons, typically between 300 and 700 Daltons. Low molecular weight compounds, in various aspects, are about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 450, about 500, about 550, about 600, about 650, about 700, about 750, about 800, about 850, about 900, or about 1000 Daltons.

METHODS

[0133] The disclosure provides compositions comprising a nanoconjugate that are able to cross the BBB. Such compositions are useful, in various aspects, for the treatment of acute and chronic disorders of the CNS. For example and without limitation, the compositions of the disclosure are useful in the treatment of acute brain and spinal cord conditions, such as focal brain ischemia, global brain ischemia, and spinal cord injury, and chronic treatment of neurodegenerative disease, including prion diseases, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), ALS, multiple sclerosis, transverse myelitis, motor neuron disease, Pick's disease, tuberous sclerosis, lysosomal storage disorders, Canavan's disease, Rett's syndrome, spinocerebellar ataxias, Friedreich's ataxia, optic atrophy, and retinal degeneration. Also contemplated for treatment are lower motor neuron diseases such as SMA and ALS as well as Pompe disease, lysosomal storage disorders, Glioblastoma multiforme and Parkinson's disease. Lysosomal storage disorders include, but are not limited to, Activator Deficiency/GM2 Gangliosidosis, Alpha-mannosidosis, Aspartylglucosaminuria, Cholesteryl ester storage disease, Chronic Hexosaminidase A Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Galactosialidosis, Gaucher Disease (Type I, Type II, Type III), GM1 gangliosidosis (Infantile, Late infantile/Juvenile, Adult/Chronic), I-Cell disease/Mucolipidosis II, Infantile Free Sialic Acid Storage Disease/ISSD, Juvenile Hexosaminidase A Deficiency, Krabbe disease (Infantile Onset, Late Onset), Metachromatic Leukodystrophy, Mucopolysaccharidoses disorders (Pseudo-Hurler

polydystrophy/Mucolipidosis IIIA, MPSI Hurler Syndrome, MPSI Scheie Syndrome, MPS I Hurler-Scheie Syndrome, MPS II Hunter syndrome, Sanfilippo syndrome Type A/MPS III A, Sanfilippo syndrome Type B/MPS III B, Sanfilippo syndrome Type C/MPS III C, Sanfilippo syndrome Type D/MPS III D, Morquio Type A/MPS INA, Morquio Type B/MPS IVB, MPS IX Hyaluronidase Deficiency, MPS VI Maroteaux-Lamy, MPS VII Sly Syndrome,

Mucolipidosis IVSialidosis, Mucolipidosis IIIC, Mucolipidosis type IV), Multiple sulfatase deficiency, Niemann-Pick Disease (Type A, Type B, Type C), Neuronal Ceroid

Lipofuscinoses (CLN6 disease (Atypical Late Infantile, Late Onset variant, Early Juvenile), Batten-Spielmeyer-Vogt/Juvenile NCL/CLN3 disease, Finnish Variant Late Infantile CLN5, Jansky-Bielschowsky disease/Late infantile CLN2/TPP1 Disease, Kufs/Adult-onset

NCL/CLN4 disease, Northern Epilepsy/variant late infantile CLN8, Santavuori- Haltia/Infantile CLN1/PPT disease, Beta-mannosidosis, Pompe disease/Glycogen storage disease type II, Pycnodysostosis, Sandhoff Disease/ Adult Onset/GM2 Gangliosidosis, Sandhoff Disease/GM2 gangliosidosis— Infantile, Sandhoff Disease/GM2 gangliosidosis— Juvenile, Schindler disease, Salla disease/Sialic Acid Storage Disease, Tay-Sachs/GM2 gangliosidosis, and/or Wolman disease. In further embodiments, use of the methods and materials is indicated for treatment of nervous system disease such as Rett Syndrome, along with nervous system injury including spinal cord and brain trauma/injury, stroke, and brain cancers.

[0134] In some aspects, a composition of the disclosure comprises a polypeptide that is a trophic or protective factor. In some embodiments, the trophic or protective factor is coadministered with a nanoconjugate of the disclosure. In various embodiments, use of a trophic or protective factor is indicated for neurodegenerative disorders contemplated herein, including but not limited to Alzheimer's Disease, Parkinson's Disease, Huntington's Disease along with nervous system injury including spinal cord and brain trauma/injury, stroke, and brain cancers. Non-limiting examples of known nervous system growth factors include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), neurotrophin-4/5 (NT-4/5), neurotrophin-6 (NT-6), ciliary neurotrophic factor (CNTF), glial cell line-derived neurotrophic factor (GDNF), the fibroblast growth factor family (e.g. , FGF's 1-15), leukemia inhibitory factor (LIF), certain members of the insulin-like growth factor family (e.g. , IGF-1), the neurturins, persephin, the bone morphogenic proteins (BMPs), the immunophilins, the transforming growth factor (TGF) family of growth factors, the neuregulins, epidermal growth factor (EGF), platelet-derived growth factor (PDGF), vascular endothelial growth factor family (e.g. , VEGF 165), follistatin, and Hifl. Also generally contemplated are zinc finger transcription factors that regulate each of the trophic or protective factors contemplated herein. In further embodiments, methods to modulate neuro- immune function are contemplated, including but not limited to, inhibition of microglial and astroglial activation through, for example, NFKB inhibition, or NFKB for neuroprotection (dual action of NFKB and associated pathways in different cell types.) by siRNA, shRNA, antisense, or miRNA. As is understood by one of skill in the art, any one or more of the aforementioned inhibitory RNAs is, in various embodiments, associated with a

nanoconjugate as described herein.

[0135] In still further embodiments, the nanoconjugate comprises a polynucleotide that specifically hybridizes to and inhibits a Bcl-2 family member. In one embodiment, the Bcl-2 family member is Bcl2L12.

[0136] In some embodiments, use of materials and methods of the disclosure is indicated for neurodegenerative disorders such as Parkinson's disease. In various embodiments, the nanoconjugate is co-administered with Aromatic acid dopa decarboxylase (AADC), Tyrosine hydroxylase, GTP-cyclohydrolase 1 (gtpchl), an apoptotic inhibitor (e.g. , bcl2, bclxL), glial cell line-derived neurotrophic factor (GDNF), the inhibitory neurotransmitter-amino butyric acid (GABA), and enzymes involved in dopamine biosynthesis. In further embodiments, the nanoconjugate is co-administered with a modifier of Parkin and/or synuclein.

[0137] In some embodiments, use of materials and methods of the disclosure is indicated for neurodegenerative disorders such as Alzheimer's disease. In further embodiments, methods to increase acetylcholine production are contemplated. In still further embodiments, methods of increasing the level of a choline acetyltransferase (ChAT) or inhibiting the activity of an acetylcholine esterase (AchE) are contemplated. [0138] In some embodiments, the nanoconjugate comprises a polynucleotide that inhibits mutant Huntington protein (htt) expression through siRNA, shRNA, antisense, and/or miRNA for treating a neurodegenerative disorder such as Huntington's disease.

[0139] In some embodiments, use of materials and methods of the disclosure is indicated for neurodegenerative disorders such as ALS. In some aspects, treatment with the embodiments contemplated by the disclosure results in a decrease in the expression of molecular markers of disease, such as TNFa, nitric oxide, peroxynitrite, and/or nitric oxide synthase (NOS).

[0140] In other aspects, the nanoconjugate comprises a short hairpin RNA directed at mutated proteins such as superoxide dismutase for ALS.

[0141] In some embodiments, use of materials and methods of the disclosure is indicated for preventing or treating neurodevelopmental disorders such as Rett Syndrome. For embodiments relating to Rett Syndrome, the nanoconjugate is co-administered with methyl cytosine binding protein 2 (MeCP2).

Methods of Inhibiting Gene Expression

[0142] Additional methods provided by the disclosure include methods of inhibiting expression of a gene product expressed from a target polynucleotide comprising contacting the target polynucleotide with a composition as described herein, wherein the contacting is sufficient to inhibit expression of the gene product. Inhibition of the gene product results from the hybridization of a target polynucleotide with a composition of the disclosure.

[0143] It is understood in the art that the sequence of a polynucleotide that is part of a nanoconjugate need not be 100% complementary to that of its target polynucleotide in order to specifically hybridize to the target polynucleotide. Moreover, a polynucleotide that is part of a nanoconjugate may hybridize to a target polynucleotide over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (for example and without limitation, a loop structure or hairpin structure). The percent complementarity is determined over the length of the polynucleotide that is part of the nanoconjugate. For example, given a nanoconjugate comprising a polynucleotide in which 18 of 20 nucleotides of the polynucleotide are complementary to a 20 nucleotide region in a target polynucleotide of 100 nucleotides total length, the polynucleotide that is part of the nanoconjugate would be 90 percent complementary. In this example, the remaining noncomplementary nucleotides may be clustered or interspersed with complementary nucleotides and need not be contiguous to each other or to complementary nucleotides. Percent complementarity of a polynucleotide that is part of a nanoconjugate with a region of a target polynucleotide can be determined routinely using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al, J. Mol. Biol., 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656).

[0144] Methods for inhibiting gene product expression provided include those wherein expression of the target gene product is inhibited by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or 100% compared to gene product expression in the absence of a nanoconjugate comprising a polynucleotide. In other words, methods provided embrace those which results in essentially any degree of inhibition of expression of a target gene product.

[0145] The degree of inhibition is determined in vivo from a body fluid sample or from a biopsy sample or by imaging techniques well known in the art. Alternatively, the degree of inhibition is determined in vitro in a cell culture assay, generally as a predictable measure of a degree of inhibition that can be expected in vivo resulting from use of a composition as described herein. It is contemplated by the disclosure that the inhibition of a target polynucleotide is used to assess the effects of the inhibition on a given cell. By way of non- limiting example, one can study the effect of the inhibition of a gene product wherein the gene product is part of a signal transduction pathway. Alternatively, one can study the inhibition of a gene product wherein the gene product is hypothesized to be involved in an apoptotic pathway.

[0146] It will be understood that any of the methods described herein can be used in combination to achieve a desired result. For example and without limitation, methods described herein can be combined to allow one to both detect a target polynucleotide as well as regulate its expression. In some embodiments, this combination can be used to quantitate the inhibition of target polynucleotide expression over time either in vitro or in vivo. The quantitation over time is achieved, in one aspect, by removing cells from a culture at specified time points and assessing the relative level of expression of a target polynucleotide at each time point. A decrease in the amount of target polynucleotide as assessed, in one aspect, through visualization of a detectable label, over time indicates the rate of inhibition of the target polynucleotide.

[0147] Thus, determining the effectiveness of a given polynucleotide to hybridize to and inhibit the expression of a target polynucleotide, as well as determining the effect of inhibition of a given polynucleotide on a cell, are aspects that are contemplated.

Use of a Nanoconjugate as a Probe

[0148] The nanoconjugates are, in one aspect, used as probes in diagnostic assays for detecting a nucleic acid or a cell.

[0149] Some embodiments of the method of detecting a target nucleic acid utilize a substrate. Any substrate can be used which allows observation of the detectable change. Suitable substrates include transparent solid surfaces (e.g. , glass, quartz, plastics and other polymers), opaque solid surface (e.g. , white solid surfaces, such as TLC silica plates, filter paper, glass fiber filters, cellulose nitrate membranes, nylon membranes), and conducting solid surfaces (e.g. , indium- tin-oxide (ITO)). The substrate can be any shape or thickness, but generally will be flat and thin. Preferred are transparent substrates such as glass (e.g. , glass slides) or plastics (e.g. , wells of microtiter plates). Methods of attaching

polynucleotides to a substrate and uses thereof with respect to nanoconjugates are disclosed in U.S. Patent Application 20020172953, incorporated herein by reference in its entirety.

[0150] By employing a substrate, the detectable change can be amplified and the sensitivity of the assay increased. In one aspect, the method comprises the steps of contacting a target polynucleotide with a substrate having a polynucleotide attached thereto, the polynucleotide (i) having a sequence complementary to a first portion of the sequence of the target nucleic acid, the contacting step performed under conditions effective to allow hybridization of the polynucleotide on the substrate with the target nucleic acid, and (ii) contacting the target nucleic acid bound to the substrate with a first type of nanoconjugate having a polynucleotide attached thereto, the polynucleotide having a sequence

complementary to a second portion of the sequence of the target nucleic acid, the contacting step performed under conditions effective to allow hybridization of the polynucleotide that is part of the nanoconjugate with the target nucleic acid. Next, the first type of nanoconjugate bound to the substrate is contacted with a second type of nanoconjugate comprising a polynucleotide, the polynucleotide on the second type of nanoconjugate having a sequence complementary to at least a portion of the sequence of the polynucleotide used to produce the first type of nanoconjugate, the contacting step taking place under conditions effective to allow hybridization of the polynucleotides on the first and second types of nanoconjugates. Finally, a detectable change produced by these hybridizations is observed.

[0151] The detectable change that occurs upon hybridization of the polynucleotides on the nanoconjugates to the nucleic acid may be a color change, the formation of aggregates of the nanoconjugates, detection of a radiological marker, or the precipitation of the aggregated nanoconjugates. The color changes can be observed with the naked eye or spectroscopically. The formation of aggregates of the nanoconjugates can be observed by electron microscopy or by nephelometry. The precipitation of the aggregated nanoconjugates can be observed with the naked eye or microscopically. Preferred are changes observable with the naked eye. Particularly preferred is a color change observable with the naked eye.

[0152] The methods of detecting target nucleic acid hybridization based on observing a color change with the naked eye are cheap, fast, simple, robust (the reagents are stable), do not require specialized or expensive equipment, and little or no instrumentation is required. These advantages make them particularly suitable for use in, e.g., research and analytical laboratories in DNA sequencing, in the field to detect the presence of specific pathogens, in the doctor's office for quick identification of an infection to assist in prescribing a drug for treatment, and in homes and health centers for inexpensive first-line screening.

[0153] A nanoconjugate comprising a polynucleotide can be used in an assay to target a target molecule of interest. Thus, the nanoconjugate comprising a polynucleotide can be used in an assay such as a bio barcode assay. See, e.g., U.S. Patent Nos. 6,361,944; 6,417,340; 6,495,324; 6,506,564; 6,582,921; 6,602,669; 6,610,491; 6,678,548; 6,677,122; 6682,895; 6,709,825; 6,720,147; 6,720,411; 6,750,016; 6,759,199; 6,767,702; 6,773,884; 6,777,186; 6,812,334; 6,818,753; 6,828,432; 6,827,979; 6,861,221; and 6,878,814, the disclosures of which are incorporated herein by reference.

[0154] In some embodiments, the compositions of the disclosure are useful in nano-flare technology. The nano-flare has been previously described in the context of polynucleotide- functionalized nanoparticles that can take advantage of a sicPN architecture for fluorescent detection of polynucleotide levels inside a living cell [described in WO 2008/098248 and U.S. Patent Application Publication Number U.S. 2011/0111974, each of which is incorporated by reference herein in its entirety]. In this system the sicPN acts as the "flare" and is detectably labeled and displaced or released from the surface by an incoming target polynucleotide. It is thus contemplated that the nano-flare technology is useful in the context of the nanoconjugates described herein.

DOSING AND PHARMACEUTICAL COMPOSITIONS

[0155] It will be appreciated that any of the compositions described herein may be administered to a mammal in a therapeutically effective amount to achieve a desired therapeutic effect.

[0156] The compositions described herein may be formulated in pharmaceutical compositions with a pharmaceutically acceptable excipient, carrier, or diluent. The compound or composition can be administered by any route that permits treatment of, for example and without limitation, a disease, disorder or infection as described herein.

Depending on the circumstances, a pharmaceutical composition is applied or instilled into body cavities, absorbed through the skin or mucous membranes, ingested, inhaled, and/or introduced into circulation. In some embodiments, a composition comprising a

nanoconjugate is administered intravenously, intraarterially, or intraperitoneally to introduce the composition into circulation. Non-intravenous administration also is appropriate, particularly with respect to low molecular weight therapeutics. In certain circumstances, it is desirable to deliver a pharmaceutical composition comprising the nanoconjugate peripherally, orally, topically, sublingually, vaginally, rectally; through injection by intracerebral (intra- parenchymal), intracerebroventricular, intramuscular, intra-ocular, intraportal, intralesional, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intranasal, urethral, or enteral means; by sustained release systems; or by implantation devices.

[0157] Administration may take the form of single dose administration, or the compound of the embodiments can be administered over a period of time, either in divided doses or in a single dose. However the compounds of the embodiments are administered to the subject, the amounts of compound administered and the route of administration chosen should be selected to permit efficacious treatment of the disease condition. Administration of combinations of therapeutic agents (i.e., combination therapy) is also contemplated, and in some of these embodiments, at least one of the therapeutic agents is in association with a nanoconjugate as described herein.

[0158] In embodiments wherein a nanoconjugate is to be studied in a glioma cell line and/or patient-derived tumor neurospheres (TNS), it is contemplated that about O. lnM to about 10 nM, or about 0.5 nM to about 8 nM, or about 1 nM to about 10 nM, or about 0.1 nM to about 0.5 nM, or about 0.1 nM to about 5 nM are administered. In specific embodiments, it is contemplated that about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nM or more of the nanoconjugate is administered to a glioma cell line and/or patient-derived TNS. In some embodiments, the administration of the nanoconjugate proceeds for about 24- 48, or about 24-36, or about 24-40, or about 36-48, or about 24, about 30, about 36, about 40, or about 48 hours or more.

[0159] In further embodiments, administration of a nanoconjugate composition as described herein is from about 1 mg/kg to about 50 mg/kg, or about 5 mg/kg to about 50 mg/kg, or from about 5 mg/kg to about 30 mg/kg, or from about 5 mg/kg to about 20 mg/kg, or from about 5 mg/kg to about 10 mg/kg. In one embodiment, the administration is intravenous administration and the amount that of the nanoconjugate composition that is administered is 7 mg/kg. In further embodiments, the nanoconjugate composition is administered daily, weekly or monthly. In some embodiments, a single administration is given per day. In further embodiments, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more administrations of a nanoconjugate composition and/or therapeutic agent are given per day, or every other day, or every week, or every month.

[0160] Administration of a nanoconjugate composition with a therapeutic agent as described herein is contemplated, in various embodiments, will begin at the same time, or the therapeutic agent will be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more days after the nanoconjugate composition. In alternative embodiments, the therapeutic agent is

administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more days before the nanoconjugate composition is administered. Therapeutically and prophylactically effective amounts of a composition for a given situation may be determined by routine experimentation that is within the skill and judgment of the clinician. For example and without limitation, the amount of temozolamide that is administered is about 10 mg/kg, or about 20 mg/kg, or about 30 mg/kg, or about 40 mg/kg, or about 50 mg/kg or more.

[0161] In an embodiment, the pharmaceutical compositions may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, and/or diluents, depending upon the particular mode of administration and dosage form. The pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5.0 to about pH 8. More particularly, the pharmaceutical compositions comprises in various aspects a therapeutically or prophylactically effective amount of at least one composition as described herein, together with one or more pharmaceutically acceptable excipients. As described herein, the pharmaceutical compositions may optionally comprise a combination of the compounds described herein.

[0162] The term "pharmaceutically acceptable excipient" refers to an excipient for administration of a pharmaceutical agent, such as the compounds described herein. The term refers to any pharmaceutical excipient that may be administered without undue toxicity.

[0163] Pharmaceutically acceptable excipients are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of

pharmaceutical compositions (see, e.g. , Remington's Pharmaceutical Sciences).

[0164] Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Other exemplary excipients include antioxidants (e.g. , ascorbic acid), chelating agents (e.g. , EDTA), carbohydrates (e.g. , dextrin, hydroxyalkylcellulose, and/or

hydroxyalkylmethylcellulose), stearic acid, liquids (e.g. , oils, water, saline, glycerol and/or ethanol) wetting or emulsifying agents, pH buffering substances, and the like.

[0165] Additionally, the pharmaceutical compositions may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous emulsion or oleaginous

suspension. This emulsion or suspension may be formulated by a person of ordinary skill in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,2-propane-diol.

[0166] The sterile injectable preparation may also be prepared as a lyophilized powder. In addition, sterile fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids (e.g. , oleic acid) may likewise be used in the preparation of injectables.

KITS

[0167] Also provided are kits comprising a composition of the disclosure. In one embodiment, the kit comprises at least one container, the container holding at least one type of nanoconjugate as described herein comprising one or more biomolecules as described herein. In aspects of the disclosure wherein the biomolecule is a polynucleotide, it is contemplated that the polynucleotides that are part of the first type of nanoconjugate have one or more sequences complementary (or sufficiently complementary as disclosed herein) to one or more sequences of a first portion of a target polynucleotide. The container optionally includes one or more additional type of nanoconjugates comprising a polynucleotide with a sequence complementary to one or more sequence of a second portion of the target polynucleotide.

[0168] In another embodiment, the kit comprises at least two containers. The first container holds one or more nanoconjugates as disclosed herein comprising one or more polynucleotides as described herein which can associate with one or more portions of a target polynucleotide. The second container holds one or more nanoconjugates comprising one or more polynucleotides that can associate with one or more sequences of the same or a different portion of the target polynucleotide.

[0169] In another embodiment, the kits have polynucleotides and nanoparticles in separate containers, and the nanoconjugates are produced prior to use for a method described herein. In one aspect, the polynucleotides and/or the nanoparticles are functionalized so that the nanoconjugates can be produced. Alternatively, the polynucleotides and/or nanoparticles are provided in the kit without functional groups, in which case they must be functionalized prior to performing the assay.

[0170] In various aspects of the kits provided, polynucleotides include a label or the kit includes a label which can be attached to the polynucleotides. Alternatively, the kits include labeled nanoparticles or labels which can be attached to the nanoparticles. In each embodiment, the kit optionally includes instructions, each container contains a label, the kit itself includes a label, the kit optionally includes one or more non-specific polynucleotides (for use as controls).

EXAMPLES

Example 1

[0171] Bcl-2-like protein 12 (Bcl2L12) expression was assessed, and the compendium of activated receptor tyrosine kinases (RTKs) in human brain tumor stem cells (BTSCs) and derived orthotopic xenografts (Figure 1). In line with previous studies in primary GBM tumor specimens [Stegh et al., Genes Dev. 21: 98-111 (2007)], robust expression of Bcl2L12 was identified in a majority of BTSCs tested and BTSC line 18 (huBTSC_18) with high Bcl2L12 expression was selected for initial functional studies (Figure 1A). In addition, it was established that multiple RTKs are co-activated in glioma cells and in their

corresponding orthotopic explants (Figure IB). Notably, the activation profile of RTKs in glioma cell lines in vitro is largely maintained in the explanted tumor - BTSC-derived grafts exhibit a more distinctive RTK signature when compared to the corresponding cultures, suggesting that the tumor microenvironment significantly impacts intratumoral RTK activation status of BTSC-initiated tumors.

Example 2

[0172] For therapeutic development, Bcl2L12 targeting RNAi gold nanoparticles (RNA- Au NP; nanoconjugates as described herein) were generated and screened for their ability to knockdown endogenous Bcl2L12 in glioma cell lines and huBTSC_18. Bcl2L12-RNA- nanoconjugates that were capable of reducing Bcl2L12 mRNA levels by 40% were identified (Figure 2A) and Bcl2L12 protein abundance by 60-95% (Figure 2B) - L12-1- and L12-2- RNA nanoconjugates. Subsequently, a nanoconjugate concentration (0.1 nM) to robustly neutralize Bcl2L12 protein expression in LN235 cells was determined (Figure 2B), which compared to 100 nM of conventional, lipoplex-delivered siRNA oligonucleotides (Figure 2C) required to achieve a similar effect, indicating that RNAi-functionalized nanoconjugates are significantly more effective in silencing gene expression than conventional methods.

Importantly, similar, highly robust KD efficacies in BTSC and confirmed persistence of Bcl2L12 protein knockdown up to 5 days post nanoconjugate-treatment was established (Figure 2D). Finally, and as shown with Bcl2L12-targeting siRNA and shRNAs [Stegh et ah, Genes Dev. 21: 98-111 (2007)], nanoconjugate-mediated knockdown of Bcl2L12 resulted in enhanced effector caspase activation as evidenced by Western Blot analyses for active caspase-3 and -7 (Figure 2E), confirming the functionality of nanoconjugate-driven Bcl2L12 knockdown.

[0173] Demonstration of knockdown of a second target: To further demonstrate the capacity of RNA- nanoconjugates to effectively silence gene expression in cells, the

Bcl2L12 downstream effector aB-crystallin (CRYAB) was selected as a second prototypic gliomagenic target. aB-crystallin is transcriptionally induced by Bcl2L12 and functions to promote tumor cell migration/invasion and inhibit effector caspase-3 activation. Figure 3 shows RNA-nanoconjugate-mediated knockdown of endogenous aB-crystallin (Figure 3A; comparison of RNA-nanoconjugate (ΙΟηΜ) and siRNA/lipoplex-mediated knockdown (100 nM)), reduced invasive properties (Figure 3B), and enhanced caspase-3 activation of glioma cells upon aB-crystallin ablation (Figure 3C). These studies demonstrated potent knockdown of two prototypic glioma oncoproteins with efficacies and impact on downstream signaling (i.e., caspase activation and cell invasion) similar to retro virally/lipolex-delivered sh/siRNAs [Stegh et al, Genes Dev. 21: 98-111 (2007); Stegh et al, Proc Natl Acad Sci U.S.A. 105: 10703-8 (2008)].

Example 3

[0174] Having established broad pro-apoptotic activities of BC12L12- and CRYAB-RNA- nanoconjugates in cell culture, the RNA-nanoconjugates' functionality in orthotopic explant and genetically engineered mice was validated in vivo. These studies tested tumor regression in a genetically engineered glioma mouse model. Expanding on the known cellular and tissue uptake properties of these nanoconjugates, penetration of RNA-nanoconjugates into normal and cancerous intracranial tissues was documented and its uptake into BTSC xenografts was assessed (Figure 4) upon IC injection. Following BTSC inoculation and Cy5- Au-NP administration, brains were dissected, and coronal sections were subjected to confocal fluorescence microscopy. Figure 4A (lower panel) shows robust dispersion of RNA- nanoconjugates within the BTSC orthotopic tumor explant similar to U87MG grafts and Figure 4B shows quantification of intracranial dispersion of fluorescence signal. Intracranial nanoconjugate uptake into tumor and non-tumor elements was verified by Inductively Coupled Plasma Mass Spectrometry (ICP-MS; Figure 4C), and also by magnetic resonance (MR) imaging using a multimodal, gadolinium (Gd(III))-enriched polyvalent DNA gold nanoparticle (DNA-Gd(III)-Au NPs) conjugate (Figure 4D).

[0175] Predominant accumulation of DNA-Gd(III)-nanoconjugates within the intracranial U87MG-xenograft is evidenced by MR and corresponding hematoxylin and eosin (H&E) images (Figure 4D, left panel). It is important to note that tumor cells and DNA-Gd(III)- nanoconjugates were injected in close proximity to Bregma; however, tumor formation and (intratumoral) accumulation of DNA-Gd(III)-nanoconjugates were most prominent in forebrain structures. This indicates that nanoconjugates migrated along the anteroposterior axis to selectively enrich tumor elements. Building on the extensive intratumoral dispersion of nanoconjugates (see Figure 4D right panel for 3D reconstruction of MR images to quantifiably assess intracranial space occupied by DNA-Gd(III)-nanoconjugates), we compared the distribution of RNA-nanoconjugates in the mouse brain using intracranial (I.C.) injection directly to the brain with systematic intravenous (I.V.) injection via tail vein. A significantly higher amount of nanoconjugates were found in the xenografted tumor than in the rest of the brain, reconfirming enhanced intratumoral accumulation of the nanoconjugates (Figure 5).

[0176] Finally, the impact of Bcl2L12-targeting RNA-nanoconjugates on tumor regression was tested in U87 glioma cell line xenogenic grafts in vivo using systemic injection. It was found that, in parallel with the in vitro findings (see Figure 2B, left panel, U87MG), mice treated with L12-2-nanoconjugate showed significantly prolonged life span (p- value = 0.01) explained by reduced activity of Bcl2L12 protein (Figure 6). LI 2-1 -nanoconjugates did not have a significant effect (p-value = 0.50) on the life span and were not effective in knockdown of Bcl2L12 in vitro in U87 cell line.

[0177] The disclosed subject matter has been described with reference to various aspects, embodiments and techniques. It should be understood, however, that many variations and modifications may be made while remaining within the spirit and scope of the disclosed subject matter. All references cited herein are hereby incorporated by reference in their entireties, or to the extent that they provide relevant disclosure, as would be ascertained from context.