Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MODULAR METAL–ORGANIC POLYHEDRA SUPERASSEMBLY COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2020/176716
Kind Code:
A1
Abstract:
A method to prepare a population of metal-organic polyhedra (MOP) supported micelle nanoparticles (NPs), and a composition comprising MOP supported micelle NPs, are provided.

Inventors:
BRINKER C JEFFREY (US)
GUO JIMIN (US)
ZHU WEI (US)
Application Number:
PCT/US2020/020066
Publication Date:
September 03, 2020
Filing Date:
February 27, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRINKER C JEFFREY (US)
GUO JIMIN (US)
ZHU WEI (US)
International Classes:
A61K9/00; A61K47/10; A61K47/34; A61P35/00; B82Y5/00; C08G83/00
Other References:
WEI ZHU, GUO JIMIN, JU YI, SERDA RITA E., CROISSANT JONAS G., SHANG JIN, COKER ERIC, AGOLA JACOB ONGUDI, ZHONG QI‐ZHI, PING YUAN, : "Modular Metal-Organic Polyhedra Superassambly: From Molecular- Level Design to Targeted Drug Delivery", ADVANCED MATERIALS, vol. 31, no. 12, 31 January 2019 (2019-01-31) - 22 March 2019 (2019-03-22), pages e1806774, XP055735218
SAMRAJ MOLLICK, SOUMYA MUKHERJEE, DONGWOOK KIM, ZHIWEI QIAO, AAMOD V. DESAI, RAJAT SAHA, YOGESHWAR D. MORE, JIANWEN JIANG, MYOUNG : "Hydrophobic Shielding of Outer Surface:Enhancing the ChemicalStability of Metal–Organic Polyhedra", ANGEWANDTE CHEMIE, vol. 58, no. 4, 21 January 2019 (2019-01-21), pages 1041 - 1045, XP055735223
MOHAMED SALMA ET AL.: "Polymeric nano-micelles: versatile platform for targeted delivery in cancer", THER. DELIV., vol. 5, no. 10, October 2014 (2014-10-01), pages 1101 - 1121
Attorney, Agent or Firm:
PERDOK, Monique M. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method to prepare a population of metal-organic polyhedra (MOP) supported micelle nanoparticles (NPs), comprising:

combining an amount of metal and an amount of an organic ligand comprising one or more hydrophobic chains under conditions to form a population of metal-organic polyhedra units; and

combining an amount of the population of metal-organic polyhedra units and an amount of a micellar solution under conditions to form single MOP-supported micelle nanoparticles (NPs).

2. The method of claim 1 wherein the metal is palladium, copper, zinc, platinum, manganese or beryllium.

3. The method of claim 1 wherein the metal is monovalent, divalent,

trivalent or tetravalent.

4. The method of claim 1, 2 or 3 wherein the hydrophobic chain is a Cl- C20 alkyl chain.

5. The method of claim 4 wherein the hydrophobic chain is a dodecyl chain.

6. The method of any one of claims 1 to 5 wherein the ligand comprises a carboxylate, such as 1,4-benzenedicarboxylic, heterocyclic azolate, pyridine, thiophene, furan, pyrrole, or cyanide.

7. The method of any one of claims 1 to 6 wherein the conditions to form single MOP-supported micelle nanoparticles comprise sonication.

8. The method of any one of claims 1 to 7 wherein the conditions to form metal-organic polyhedra units yield a precipitate.

9. The method of any one of claims 1 to 8 wherein the conditions to form metal-organic polyhedra units comprise applying heat.

10. The method of any one of claims 1 to 9 wherein the diameter of the metal-organic polyhedra unit is from about 1 nm to about 15 nm. 11. The method of any one of claims 1 to 9 wherein the diameter of the

metal-organic polyhedra unit is from about 10 nm to about 25 nm.

12. The method of any one of claims 1 to 9 wherein the diameter of the

metal-organic polyhedra unit is from about 3 nm to about 10 nm.

13. The method of any one of claims 1 to 12 wherein the diameter of the micelle is from about 10 nm to about 25 nm, 10 nm to about 20 nm or about 15 nm to about 25 nm. 14. The method of any one of claims 1 to 12 wherein the diameter of the micelle is about 100 nm to about 800 nm, about 200 nm to about 500 nm or about 500 nm to about 900 nm.

15. The method of any one of claims 1 to 12 wherein the diameter of the micelle is from about 1000 nm to about 3000 nm or about 1500 nm to about 2500 nm.

16. The method of any one of claims 1 to 15 wherein the micelle solution comprises PEG, DSPE, or combination thereof.

17. The method of claim 16 wherein the PEG has 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 PEO units.

18. The method of any one of claims 1 to 17 further comprising forming superassemblies of the micelle.

19. The method of claim 18 wherein the diameter of the superassemblies is from about 100 nm to about 900 nm. 20 The method of claim 18 wherein the diameter of the superassemblies is from about 100 nm to about 300 nm.

21. The method of claim 18 wherein the diameter of the superassemblies is from about 300 nm to about 500 nm, 400 nm to about 600 nm or 600 nm to about 800 nm.

22. The method of claim 18 wherein the diameter of the superassemblies is from about 1 pm to about 100 pm or 1 pm to about 10 pm.

23. The method of any one of claims 1 to 22 wherein the MOP-supported micelle nanoparticles or superassemblies further comprise one or more cargo molecules. 24. The method of claim 23 wherein the one or more cargo molecules

comprise a drug, a dye or a contrast agent.

25. The method of claim 23 wherein the one or more cargo molecules

comprise an antibody or a fragment thereof, a protein ligand, a quantum dot or a gold nanoparticle.

26. The method of any one of claims 1 to 25 wherein the MOP-supported micelle nanoparticles or superassemblies comprise one or more targeting molecules.

27. A population of single MOP-supported micelle nanoparticles prepared by the method of any one of claims 1 to 26.

28. A population of superassemblies of metal-organic polyhedral prepared by the method of any one of claims 18 to 27.

29. A method to prevent, inhibit or treat cancer in a mammal, comprising: administering to the mammal an effective amount of a composition comprising the population of claim 27 or 28 which comprises one or more chemotherapeutic drugs.

30. The method of claim 29 wherein the mammal is a human.

31. The methods of claim 29 or 30 wherein the composition is injected.

32. The methods of claim 29 or 30 wherein the composition is locally

administered.

33. The methods of claim 29 or 30 wherein the composition is systemically administered.

34. The method of claim 33 wherein the composition is intravenously

administered.

35. A method to deliver a drug to a mammal, comprising: administering to the mammal an effective amount of a composition comprising the population of claim 27 or 28 which comprises one or more drugs.

36. A method for imaging in a mammal, comprising: administering to the mammal an effective amount of a composition comprising the population of claim 27 or 28 which comprises one or more imaging agents. 37. Use of the population of claim 27 or 28.

38. A support comprising the population of claim 27 or 28.

39. The support of claim 38 which is a membrane.

40. The support of claim 39 wherein the membrane is a polypropylene, cellulose acetate, cellulose nitrate, cellulose ester, nylon,

polyethersulfone, polyester, polypropylene, or polycarbonate membrane.

41. A micelle comprising a plurality of metal-organic polyhedral units having metal nodes and an organic ligand comprising one or more hydrophobic chains. 42. The micelle of claim 41 wherein the diameter of the metal-organic polyhedra unit is from about 1 nm to about 15 nm.

43. The micelle of claim 41 wherein the diameter of the metal-organic polyhedra unit is from about 10 nm to about 25 nm.

44. The micelle of any one of claims 41 to 43 which comprises PEG.

45. The micelle of any one of claims 41 to 43 which comprises one or more drugs or imaging agents.

Description:
MODULAR METAL-ORGANIC POLYHEDRA

SUPERASSEMBLY COMPOSITIONS

Cross-Reference to Related Applications

This application claims the benefit of the filing date of U.S. application No. 62/811,668, filed on February 28, 2019, the disclosure of which is incorporated by reference herein.

Statement of Government Rights

This invention was made with government support under FP0003261 awarded by the National Institutes of Health and DE-NA0003525 awarded by the Department of Energy. The government has certain rights in the invention.

Background

The desire to improve drug efficacy, enhance targeted delivery to specific sites, and reduce side effects by controlling drug pharmacokinetic and biodistribution profiles has remained the main driving force for the development of drug delivery systems over the past two decades (Mitragotri et al, 2014; Blanco et al., 2015). Among all of the developed nanocarriers (e.g., micelles, liposomes, and mesoporous silica nanoparticles (NPs)) (Elsabahy et al, 2015; Grim et al., 2016; Croissant et al., 2017; Zhu et al., 2018; Richardson et al, 2016), nanosized metal-organic polyhedra (MOP) obtained upon metal-ligand coordination have attracted increasing attention for drug delivery owing to the highly designable nanoarchitectures, well-defined pore cavities, as well as diverse chemical properties and functions (Cook et al, 2013; Ahmad et al,

2015; Harris et al., 2013; Grishagin et al, 2014; Yu et al, 2016; Varhan et al, 2016). In this field, Therrien et al. presented a series of organometallic cages as anticancer drug delivery vehicles for photodynamic therapy (Therrien et al., 2008; Schmitt et al, 2012). Lippard and co-workers reported a well-defined metal-organic octahedron that can enhance the delivery of cis- platin prodrugs to cancer cells (Zheng et al., 2015). In addition, Isaacs and co-workers highlighted the potential of host-guest interactions, by combining MOP with cucurbituril, for enhanced delivery of chemotherapeutic drugs (Samanta et al, 2015; Samanta et al., 2017). Although many MOP -based supramolecular systems have been developed for drug delivery applications (Zhao et al, 2011; Xu et al., 2017; Rodriguez et al, 2017; Croissant et al, 2018), compared to the well-developed NP -based systems such as mesoporous silica NPs (Croissant et al, 2018), the use of MOP for drug delivery is still in its infancy. This is especially the case IN research concerning targeted drug delivery for cancer therapy. A major limitation of MOP as nanocarriers is their rapid renal clearance and short circulation time owing to their small size, which is typically below the filtration barrier of the glomerulus (e.g., ~5.5 nm) (Samanta et al., 2016; Samanta et al, 2017). Furthermore, using coordination complexes for targeted therapy requires their functionalization with various targeting ligands consisting of small molecular moieties or large antibodies.

Summary

Targeted drug delivery remains at the forefront of biomedical research but remains a challenge to date. To overcome these limitations, the

superassembly of nanosized metal-organic polyhedra (MOP) and their biomimetic coatings of lipid bilayers are described herein, which synergistically combines the advantages of micelles and supramolecular coordination cages for targeted drug delivery. The superassembly technique affords unique

hydrophobic features that endow individual MOP to act as nanobuilding blocks and enable their superassembly into larger well-defined nanocarriers with homogeneous sizes over a broad range of diameters. Various cargos are controllably loaded into the MOP with high payloads, and the nanocages are then superassembled to form multidrug delivery systems. Additionally, functional nanoparticles are introduced into the superassemblies via a one-pot process for versatile bioapplications. The MOP superassemblies are surface- engineered with epidermal growth factor receptors and can be targeted to cancer cells. In vivo studies indicated the assemblies have a substantial circulation half- life of 5.6 h and undergo renal clearance, characteristics needed for

nanomedicines.

In one embodiment, a method to prepare a population of metal-organic polyhedra (MOP) supported micelle nanoparticles (NPs) is provided. The method includes combining metal to form nodes and an organic ligand comprising one or more hydrophobic chains under conditions to form a population of metal-organic polyhedra units; and combining the population of metal-organic polyhedra units and a micellar solution under conditions to form single MOP-supported micelle nanoparticles (NPs). In one embodiment, the metal is palladium, copper, zinc, platinum, manganese, beryllium, iron, chromium, cobalt, aluminum, zirconium, indium, or europium.. In one embodiment, the metal is monovalent, divalent, trivalent or tetravalent. In one embodiment, the hydrophobic chain is a C1-C20 alkyl chain. In one

embodiment, the hydrophobic chain is a dodecyl chain. In one embodiment, the hydrophobic chain is a octyl chain. In one embodiment, the organic ligand comprises a carboxylate, such as 1,4-benzenedicarboxylic, heterocyclic azolate, pyridine, thiophene, furan, pyrrole, or cyanide.

In one embodiment, the conditions to form single MOP-supported micelle nanoparticles comprise sonication. In one embodiment, the conditions to form metal-organic polyhedra units comprise applying heat. In one

embodiment, the conditions to form metal-organic polyhedral yield a precipitate. In one embodiment, the diameter of a metal-organic polyhedra unit is from about 1 nm to about 15 nm. In one embodiment, the diameter of the metal-organic polyhedra unit is from about 10 nm to about 25 nm. In one embodiment, the diameter of the metal-organic polyhedra unit is from about 3 nm to about 10 nm. In one embodiment, the diameter of the micelle is from about 5 nm to about 10 nm. In one embodiment, the diameter of the micelle is from about 10 nm to about 25 nm, 10 nm to about 20 nm or about 15 nm to about 25 nm. In one embodiment, the diameter of the micelle is about 100 nm to about 800 nm, about 200 nm to about 500 nm or about 500 nm to about 900 nm. In one embodiment, the diameter of the micelle is from about 1000 nm to about 3000 nm or about 1500 nm to about 2500 nm. In one embodiment, the micelle solution comprises PEG, DSPE, or combination thereof. In one embodiment, the micelle solution comprises DSPC, DSPE, DSPE-PEG, DSPE-PEG-biotin, DSPE-PEG-carboxy NHS, DPPC, DPPE, DMPC, DOPC, DOPE, DOPG, DOPS, DOTAP, DOPE- PEG-amine, DOPE-PEG-azide, or a combination thereof. In one embodiment, the PEG has a chain length (number of PEO units) of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25. In one embodiment, the PEG has a chain length (number of PEO units) of 6, 9, 10, 23, 34, 45, 68, 90, 113,

136, 181, or 227. In one embodiment, the method further comprises forming superassemblies with the MOP containing micelle. In one embodiment, the diameter of the superassemblies is from about 100 nm to about 900 nm. In one embodiment, the diameter of the superassemblies is from about 100 nm to about 300 nm. In one embodiment, the diameter of the superassemblies is from about 300 nm to about 500 nm, 400 nm to about 600 nm or 600 nm to about 800 nm.

In one embodiment, the diameter of the superassemblies is from about 1 pm to about 100 pm or 1 pm to about 10 pm. In one embodiment, the diameter of the superassemblies is from about 100 pm to about 300 pm. In one embodiment, the diameter of the superassemblies is from about 300 pm to about 500 pm, 400 pm to about 600 pm or 600 pm to about 800 pm. In one embodiment, the MOP containing micelles or superassemblies further comprise one or more cargo molecules. In one embodiment, the cargo comprises a drug, a dye or a contrast agent, or combinations thereof. In one embodiment, the cargo comprises an antibody or a fragment thereof, a protein ligand, a quantum dot or a gold nanoparticle. In one embodiment, the MOPs in the superassemblies have more than one type of drug, cell, contrast or imaging agent, protein or nanoparticle. In one embodiment, the micelle or superassemblies comprise one or more targeting molecules. Populations of the single MOP-supported micelle nanoparticles and of the superassemblies, and uses thereof are, also provided. In one embodiment, one or more different cargo molecules are mixed with metal-organic polyhedra, then with micelles to form superassemblies. In one embodiment, different ratios of cargo loaded metal-organic polyhedral and micelles are mixed. In one embodiment, a protein or other ligand, e.g., antibody, is grafted onto the surface of the micelle. In one embodiment, a population of superassemblies is added to a membrane. In one embodiment, a drug, e.g., doxorubicin, cyclophosphamide, gemcitabine, cytarabine, paclitaxel, docetaxel, vincristine sulfate, afatinib, dexamethasone, or rapamycin, are loaded into the polyhedral.

Brief Description of the Figures

Figures lAa-lAb. Schematic illustration of a) the design and

construction of drug delivery nanocarriers (single MOP or MOP superassembly- supported micelle) based on a modular superassembly approach and b) the solution synthesis of MOF NP-supported lipid bilayers based on MBB approach.

Figures IBa-lBf. a) Self-assembly of Pd24L48-Ci2 MOP with

hydrophobic chain decoration and lateral fusion of micelles b) 'HNMR spectra of free ligand and Pd 24 L 48 -Ci 2 MOP in DMSO-d 6 solvent c) HR-TEM image of the Pd24l-48-Ci2 MOP assembly; the distance between two closed packed cages is also highlighted d) Optimized structures of Pd 24 L 48 -Ci 2 MOP based on MM calculations e) Dark-field STEM image of single Pd 24 L 48 -Ci 2 MOP-supported micelle and the related size distribution f) DLS data of PeLEL-based micelle before and after loading with single Pd 24 L 48 -Ci 2 MOP NPs.

Figures 2A-2G. A) Schematic illustration of the construction of single MOP@micelle and MOP Sa @rnicelles with different size scales. B) DLS data of Pd24L48-Ci2 MOP-supported micelles with ultrasmall, nanometer, and micrometer sizes. C) DLS data showing size control of MOPsa@micelles. D) AFM images of MOP Sa @micelles with nanometer size. E,F) HR-TEM images and G) simulated structure of the dense packing of Pd24L48-Ci2 MOP in the superassembly.

Figures 3A-3F. A) UV-vis spectra of free DOX, free Pd24L48-Ci2 MOP, and DOX@Pd 24 L 48 -Ci 2 MOP in DMSO solution. B) UV-vis spectra of the multiple dye-loaded MOP sa @rnicelles with different dye loading ratios. C) UV- vis and emission spectra of CdSe/ZnS QDs@MOP sa @ micelles and UV-vis spectrum of Au NPs@MOP Sa @rnicelles. The insets show the corresponding optical fluorescent and optical images. D) Schematic illustration of the fabrication of MOP superassembly-based separation membrane. E) SEM images of the MOP superassembly -based coating on a porous polypropylene-based substrate. F) The use of Pd 24 L 48 -Ci 2 MOP superassembly -based membrane for sulforhodamine B separation.

Figures 4A-4E. A) Percent hemolysis and B) photographs of human RBCs incubated with single MOP-supported micelle and MOP superassembly- supported micelles with different size scales. C) Long-term colloidal stability of various MOP superassembly-supported micelles in different media: PBS, DMEM, and F-12K at 25°C. D) Time-dependent drug (DOX) release behavior of MOP superassembly-supported micelles in PBS solution at different pH values. E) Cytotoxicity profiles of single MOP-supported micelle and MOP superassembly-supported micelles with different PEG chain lengths (size: -162 nm) and sizes (PEG23-based) against A549 and HeLa cells.

Figures 5A-5F. A) Schematic illustration of the construction of EGFR- modified MOP Sa @rnicelle for targeted drug delivery. Fluorescence microscopy images of A549 cells treated with MOP sa @micelles B) without and C) with EGFR modification after incubation for 45 minutes at 37°C. Flow cytometry analysis of A549 cancer cells incubated with red fluorescent dye-loaded

MOP sa @micelles D) without or E) with EGFR modification at multiple time points. F) Sustained viability of A549 cells after incubation of MOP Sa @micelle with or without EGFR modification or DOX loading for 1 hour.

Figure 6. Fourier transform infrared spectrophotometry (FT-IR) of organic ligand and the formed Pd24L48-Ci2 MOP.

Figure 7. Electrospray ionization mass spectrometry pattern of the molecular cage Pd24L48 with no hydrophobic alkane chains decoration.

Figure 8. AFM image of the molecular cage Pd 24 L 48 -Ci 2 MOP on silicon substrate.

Figure 9. AFM images of MOP Sa @micelles with different sizes.

Figure 10. Ar sorption isotherm of the MOP Sa @micelle.

Figure 11. Optimized structure of Pd24L48-Ci2 MOP based on molecular mechanics calculation with a relative large pore window of 1.4 nm x 1.4 nm.

Figure 12A-12B. UV-Vis spectra of the dye-loaded Pd24L48-Ci2 MOP and free dyes: (a) sulforhodamine B and (b) Mn-TCPP.

Figure 13. TEM image of DOX-loaded MOP Sa @micelle.

Figures 14A-14B. TEM image of the commecial CdSe/ZnS quantum dots (A) and the synthesized Au NPs (B).

Figure 15. TEM image of the Au NPs@MOP Sa @micelle. The Au NPs were pointed out by arrows.

Figure 16. Fluorescent image of the CdSe/ZnS quantum dot@ MOP sa @ micelles with fluorescein isothiocyanate dye previously loaded in MOP nanocavities. The overlapping of the fluorescent points from different channels confirms the successful doping of quantum dots in MOP Sa @micelles.

Figures 17A-17D. Cytotoxicity profiles of single MOP-supported micelle and MOP super-assembly-supported micelles with different PEG chain lengths and sizes against (A,B) A549 and (C,D) HeLa cells.

Figure 18. Sustained viability of A549 cells after incubation of free DOX, MOP sa @micelle without EGFR modification, and DOX-loaded

MOP sa @micelle without EGFR modification for 2 hours.

Figure 19. The circulation of the created MOP sa @ micelle NPs in mice. Figure 20. Semilog plot of the circulation of the created MOP sa @ micelle NPs in mice.

Figure 21. Fluorescence images of different organs at 6, 12, 24, and 48 hours after intravenous administration of the MOP sa @ micelle NPs.

Figure 22. Fluorescence intensity per gram of tissue at 6, 12, 24, and 48 hours after intravenous administration of the MOPsa@ micelle NPs.

Detailed Description

The disclosed fabrication technique affords a synthesis process and the organization of individual MOP to form advanced hierarchical structures.

Specifically, in one embodiment, the superassembly of MOP supported micelles (MOPsa@micelle) for targeted drug delivery is described (see Scheme la in Figure 1A). The decoration of the outer surface of the MOP with hydrophobic chains transforms MOP units into nanobuilding blocks that can self-assemble into larger and well-defined superassemblies within micelles. This approach is different from the reported molecular building block (MBB) approach that is used to construct metal-organic framework (MOF)-based lipid bilayers, where the open metal sites of MOF units allow further coordination of additional organic ligands to form MOF NPs (Scheme lb in Figure 1 A). Nonetheless, the MBB approach typically affords limited size control, whereas the superassembly approach described herein provides homogeneous size distributions in broad ranges of diameters through the simple control of the MOP concentration to obtain ultrasmall single MOP@micelle and MOPsa@micelle. In addition, owing to the highly porous nature of the MOP, various molecules including cancer drugs can be controllably loaded into individual MOP units with high payloads and subsequently integrated into MOP superassemblies to form multiple compound (cargo), such as multiple drug, delivery systems. Furthermore, the MOP superassembly approach enabled the modular assembly of MOPs with additional functional NPs such as fluorescent quantum dots (QDs) for multifluorescence imaging or gold NPs for enhanced bioimaging.

To demonstrate the potential of the MOP superassembly concept for drug delivery, a Fujita-type MOP composed of dozens of dodecyl chains was chosen as a prototype. As described herein below, a dose-dependent assay for MOPsa@ micelles of different sizes demonstrated negligible hemolytic activities and long- term colloidal stability in various media, thereby mitigating possible concerns of structure disassembly during circulation. Cell viability tests further demonstrated the good biocompatibility of the MOPsa@micelles that is related to the NP size and the molecular weight of the polyethylene glycol (PEG) that was used for surface modification. Importantly, designing MOPsa@micelle nanocarriers with targeting moieties enabled targeted cell, e.g., targeted cancer cell, delivery and in vivo experiments using a mouse model also confirmed the good circulation. Taken together, this modular superassembly approach combines the synergistic advantages of micelles (e.g., low inherent toxicity and long circulation time) and the MOP superassembly (e.g., highly controlled architecture, stability, and high payloads of multiple cargos), and promotes the design of MOP-inspired nanocarriers for targeted cancer therapies.

The MOP unit may contain any metal including but not limited to Pd, Pt, Be, Cu, Zn, Ag, Mg, Mn, Fe, Co, Ni, Cd, Al, Sc V, Cr, Ga, In, lanthanide, Ti, Zr, Hf, Rh, or Ce, or any combination thereof, and the polyhedra may be of any shape, including but not limited to a tetrahedron, octahedron, cube, icosahedron, dodecahedron, tricontahedron,

icosadodecahedron, rhomic dodecahedron or cub -octahedron

Any ligand may be employed in the synthetic method, ligands including ligand 1 in Figure IB, the ligand in Samanta et al. (2016), the ligand in Samanata et al. (2018), and those in the Figures in Vardhan and Verpoort (Aust. J. Chem.. 68:707 (2015), the disclosures of which are incorporated by reference herein.

The hydrophobic chain that may be employed with the ligand includes but is not limited to a substituted or unsubstituted alkyl chain, e.g., a C3 to CIO, C12 to C20 or C20 to C30 alkyl chain, a substituted or un substituted alkyene chain e.g., a C3 to CIO, C12-C20,or C20 to C30 alkenyl chain, or a substituted or unsubstituted alkyne chain. When heteroatoms (N, O and S typically) are allowed to replace carbon atoms as in heteroalkyl groups, for example, the numbers describing the group, though still written as e.g. C1-C6, represent the sum of the number of carbon atoms in the group plus the number of such heteroatoms, that are included as replacements for carbon atoms in the backbone of the ring or chain being described. Alkyl, alkenyl and alkynyl groups are often optionally substituted to the extent that such substitution makes sense chemically. Typical substituents include, but are not limited to, halo, =0, =N-CN, =N-OR, =NR, OR, NR 2 , SR, SO 2 R, SO2NR2, NRSO2R, NRCONR2, NRCOOR, NRCOR, CN, COOR, COMO, OOCR, COR, and NO 2 , wherein each R is independently H, C i-Cx alkyl, C 2 -C 8 heteroalkyl, Ci-Cs acyl, C 2 -C 8 heteroacyl, C 2 -C 8 alkenyl, C 2 -C 8 heteroalkenyl, C 2 -C 8 alkynyl, C 2 -C 8 heteroalkynyl, C 6 -C 10 aryl, or C 5 -C 10 heteroaryl, and each R is optionally substituted with halo, =0, =N-CN, =N-OR’, =NR’, OR’, Ml’ 2 , SR’, S0 2 R\ S0 2 NR’2, NR’SChR’, NR’CONR’2, NR’ COOR’, NR’ COR’, CN, COOR’, COMC 2 , OOCR’, COR’, and NO 2 , wherein each R’ is independently H, Ci-Cs alkyl, C2-C8 heteroalkyl, Ci-Cs acyl, C2-C8 heteroacyl, C6-C10 aryl or C 5 -C 10 heteroaryl. Alkyl, alkenyl and alkynyl groups can also be substituted by Ci-Cs acyl, C 2 -C 8 heteroacyl, C 6 -C 10 aryl or C 5 -C 10 heteroaryl, each of which can be substituted by the substituents that are appropriate for the particular group.

“Heteroalkyl”,“heteroalkenyl”, and“heteroalkynyl” and the like are defined similarly to the corresponding hydrocarbyl (alkyl, alkenyl and alkynyl) groups, but the‘hetero’ terms refer to groups that contain one to three O, S or N heteroatoms or combinations thereof within the backbone residue; thus at least one carbon atom of a corresponding alkyl, alkenyl, or alkynyl group is replaced by one of the specified heteroatoms to form a heteroalkyl, heteroalkenyl, or heteroalkynyl group. The typical sizes for heteroforms of alkyl, alkenyl and alkynyl groups are generally the same as for the corresponding hydrocarbyl groups, and the substituents that may be present on the heteroforms are the same as those described above for the hydrocarbyl groups. For reasons of chemical stability, it is also understood that, unless otherwise specified, such groups do not include more than two contiguous heteroatoms except where an oxo group is present on N or S as in a nitro or sulfonyl group.

While“alkyl” as used herein includes cycloalkyl and cycloalky lalkyl groups, the term“cycloalkyl” may be used herein to describe a carbocyclic non aromatic group that is connected via a ring carbon atom, and“cycloalkylalkyl” may be used to describe a carbocyclic non-aromatic group that is connected to the molecule through an alkyl linker. Similarly,“heterocyclyl” may be used to describe a non-aromatic cyclic group that contains at least one heteroatom as a ring member and that is connected to the molecule via a ring atom, which may be C or N; and“heterocyclylalkyl” may be used to describe such a group that is connected to another molecule through a linker. The sizes and substituents that are suitable for the cycloalkyl, cycloalky lalkyl, heterocyclyl, and

heterocyclylalkyl groups are the same as those described above for alkyl groups. As used herein, these terms also include rings that contain a double bond or two, as long as the ring is not aromatic.

As used herein,“acyl” encompasses groups comprising an alkyl, alkenyl, alkynyl, aryl or ary lalkyl radical attached at one of the two available valence positions of a carbonyl carbon atom, and heteroacyl refers to the corresponding groups wherein at least one carbon other than the carbonyl carbon has been replaced by a heteroatom chosen from N, O and S. Thus heteroacyl includes, for example, -C(=0)OR and -C(=0)NR 2 as well as -C(=0)-heteroaryl.

Acyl and heteroacyl groups are bonded to any group or molecule to which they are attached through the open valence of the carbonyl carbon atom. Typically, they are Ci-Cs acyl groups, which include formyl, acetyl, pivaloyl, and benzoyl, and C2-C8 heteroacyl groups, which include methoxyacetyl, ethoxycarbonyl, and 4-pyridinoyl. The hydrocarbyl groups, aryl groups, and heteroforms of such groups that comprise an acyl or heteroacyl group can be substituted with the substituents described herein as generally suitable substituents for each of the corresponding component of the acyl or heteroacyl group.

“Aromatic” moiety or“aryl” moiety refers to a monocyclic or fused bicyclic moiety having the well-known characteristics of aromaticity; examples include phenyl and naphthyl. Similarly,“heteroaromatic” and“heteroaryl” refer to such monocyclic or fused bicyclic ring systems which contain as ring members one or more heteroatoms selected from O, S and N. The inclusion of a heteroatom permits aromaticity in 5 membered rings as well as 6 membered rings. Typical heteroaromatic systems include monocyclic C5-C6 aromatic groups such as pyridyl, pyrimidyl, pyrazinyl, thienyl, furanyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, and imidazolyl and the fused bicyclic moieties formed by fusing one of these monocyclic groups with a phenyl ring or with any of the heteroaromatic monocyclic groups to form a Cs-Cio bicyclic group such as indolyl, benzimidazolyl, indazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, pyrazolopyridyl, quinazolinyl, quinoxalinyl, cinnolinyl, and the like. Any monocyclic or fused ring bicyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system is included in this definition. It also includes bicyclic groups where at least the ring which is directly attached to the remainder of the molecule has the characteristics of aromaticity. Typically, the ring systems contain 5-12 ring member atoms. For example, the monocyclic heteroaryls may contain 5-6 ring members, and the bicyclic heteroaryls contain 8-10 ring members.

Aryl and heteroaryl moieties may be substituted with a variety of substituents including Ci-Cs alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C5-C12 aryl, Ci- C 8 acyl, and heteroforms of these, each of which can itself be further substituted; other substituents for aryl and heteroaryl moieties include halo, OR, NR2, SR, SO2R, SO2NR2, NRSO2R, NRCONR2, NRCOOR, NRCOR, CN, COOR, CONR2, OOCR, COR, and NO2, wherein each R is independently H, Ci-Cs alkyl, C2-C8 heteroalkyl, C2-C8 alkenyl, C2-C8 heteroalkenyl, C2-C8 alkynyl, C2- C 8 heteroalkynyl, C6-C10 aryl, C5-C10 heteroaryl, C7-C12 arylalkyl, or C6-Ci2 heteroarylalkyl, and each R is optionally substituted as described above for alkyl groups. The substituent groups on an aryl or heteroaryl group may of course be further substituted with the groups described herein as suitable for each type of such substituents or for each component of the substituent. Thus, for example, an arylalkyl substituent may be substituted on the aryl portion with substituents described herein as typical for aryl groups, and it may be further substituted on the alkyl portion with substituents described herein as typical or suitable for alkyl groups.

Components of the micelle include but are not limited to any lipid or polymer including but not limited to l,2-dioleoyl-.s//-glycero-3-phosphocholine (DOPC), 1 , 2-dipalm itoyl-.s//-glycero-3-phosphocholine (DPPC), 1,2-distearoyl- ST?-glycero-3-phosphocholine (DSPC), l,2-dioleoyl-sn-glycero-3-[phosphor-L- serine] (DOPS), l,2-dioleoyl-3-trimethylammonium-propane (18: 1 DOTAP), l , 2-dioleoyl-v // -glycero-3-phospho-(r- / .-glycerol) (DOPG), 1 , 2-dioleoyl-.s // - glycero-3-phosphoethanolamine (DOPE), 1, 2-dipalm itoyl-.s//-glycero-3- phosphoethanolamine (DPPE), l,2-dioleoyl-.s//-glycero-3-phosphoethanolamine- N-[methoxy(polyethylene glycol)-2000] (18: 1 PEG-2000 PE), 1,2-dipalmitoyl- s«-glycero-3-phosphoethanolamine-N-[methoxy(poly ethylene glycol)-2000] (16:0 PEG-2000 PE), l-oleoyl-2-[12-[(7-nitro-2-l,3-benzoxadiazol-4- yl)amino]lauroyl]-s«-glycero-3-phosphocholine (18:1-12:0 NBD PC), 1- palmitoy 1-2- ( 12- [(7-nitro-2- 1 ,3 -b enzoxadiazol-4-y l)am inojlauroy 1 } -s//-glycero- 3-phosphocholine (16:0-12:0 NBD PC), cholesterol and mixtures/combinations thereof. Cholesterol, not technically a lipid, but presented as a lipid for purposes of an embodiment of the given the fact that cholesterol may be an important component of the lipid bi-layer of protocells according to an embodiment. Often cholesterol is incorporated into lipid bi-layers of protocells in order to enhance structural integrity of the bi-layer. These lipids are all readily available commercially from Avanti Polar Lipids, Inc. (Alabaster, Alabama, USA).

DOPE and DPPE are particularly useful for conjugating (through an appropriate crosslinker) peptides, polypeptides, including antibodies, RNA and DNA through the amine group on the lipid.

Ion one embodiment, components of the micelle include but are not limited

to DSPC, DSPE, DSPE-PEG, DSPE-PEG-biotin, DSPE-PEG-carboxy NHS, DPPC, DPPE, DMPC, DOPC, DOPE, DOPG, DOPS, DOTAP, DOPE-PEG- amine, DOPE-PEG-azide, or a combination thereof.

In certain embodiments, the micelle is comprised of one or more phosphatidyl-cholines (PCs) selected from the group consisting of 1,2- distearoyl-.s//-glycero-3-phosphocholine (DSPC) [18:0], l ,2-dioleoyl-.s//-glycero- 3-phosphocholine (DOPC) [18: 1 (A9-Cis)], l,2-dimyristoyks77-glycero-3- phosphocholine (DMPC), l,2-dioleoyl-3-trimethylammonium-propane

(DOTAP), l -palmitoyl-2-oleoyl-.s//-glycero-3-phosphocholine (POPC), egg PC, and a lipid mixture comprising of one or more unsaturated phosphatidyl cholines, DMPC [14:0] having a carbon length of 14 and no unsaturated bonds, l,2-dipalmitoyks77-glycero-3-phosphocholine (DPPC) [16:0], POPC [16:0-18:1], and DOTAP [18: 1] The use of DSPC and/or DOPC as well as other

zwitterionic phospholipids as a principal component (often in combination with a minor amount of cholesterol) is employed in certain embodiments in order to provide a protocell with a surface zeta potential which is neutral or close to neutral in character. In other embodiments, the micelle is comprised of a mixture of (1)

DSPC, DOPC and optionally one or more phosphatidyl-cholines (PCs) selected from the group consisting of l ,2-dimyristoyl-.s//-glycero-3-phosphocholine (DMPC), 1, 2-dioleoy 1-3 -trim ethylammonium-propane (DOTAP), l-palmitoyl-2- oleoyl-.s//-glycero-3-phosphocholine (POPC), a lipid mixture comprising (in molar percent) between about 50% to about 70% or about 51% to about 69%, or about 52% to about 68%, or about 53% to about 67%, or about 54% to about 66%, or about 55% to about 65%, or about 56% to about 64%, or about 57% to about 63%, or about 58% to about 62%, or about 59% to about 61%, or about 60%, of one or more unsaturated phosphatidyl-choline, DMPC [14:0] having a carbon length of 14 and no unsaturated bonds, l,2-dipalmitoyl-s«-glycero-3- phosphocholine (DPPC) [16:0], POPC [16:0-18:1] and DOTAP [18:1]; and wherein (b) the molar concentration of DSPC and DOPC in the mixture is between about 10% to about 99% or about 50% to about 99%, or about 12% to about 98%, or about 13% to about 97%, or about 14% to about 96%, or about 55% to about 95%, or about 56% to about 94%, or about 57% to about 93%, or about 58% to about 42%, or about 59% to about 91%, or about 50% to about 90%, or about 51% to about 89%.

In certain embodiments, the micelle is comprised of one or more compositions selected from the group consisting of a phospholipid, a

phosphatidyl-choline, a phosphatidyl-serine, a phosphatidyl-diethanolamine, a phosphatidylinosite, a sphingolipid, and an ethoxylated sterol, or mixtures thereof. In illustrative examples of such embodiments, the phospholipid can be a lecithin; the phosphatidylinosite can be derived from soy, rape, cotton seed, egg and mixtures thereof; the sphingolipid can be ceramide, a cerebroside, a sphingosine, and a sphingomyelin, and a mixture thereof; the ethoxylated sterol can be phytosterol, PEG-(polyethyleneglycol)-5-soy bean sterol, and PEG- (polyethyleneglycol)-5 rapeseed sterol. In certain embodiments, the phytosterol comprises a mixture of at least two of the following compositions: sitosterol, campesterol and stigmasterol.

In still other illustrative embodiments, the micelle is comprised of one or more phosphatidyl groups selected from the group consisting of phosphatidyl choline, phosphatidyl-ethanolamine, phosphatidyl-serine, phosphatidyl- inositol, lyso-phosphatidy 1-choline, lyso-phosphatidyl-ethanolamine, lyso-phosphatidyl- inositol and lyso-phosphatidyl-inositol.

In still other illustrative embodiments, the micelle is comprised of phospholipid selected from a monoacyl or diacylphosphoglyceride.

In still other illustrative embodiments, the micelle is comprised of one or more phosphoinositides selected from the group consisting of phosphatidyl- inositol-3-phosphate (PI-3-P), phosphatidyl-inositol-4-phosphate (PI-4-P), phosphatidyl-inositol-5-phosphate (PI-5-P), phosphatidyl-inositol-3, 4- diphosphate (PI-3, 4-P2), phosphatidyl-inositol-3, 5-diphosphate (PI-3, 5-P2), phosphatidyl-inositol-4, 5-diphosphate (PI-4, 5-P2), phosphatidyl-inositol-3, 4,5- triphosphate (PI-3,4,5-P3), lysophosphatidyl-inositol-3 -phosphate (LPI-3-P), lysophosphatidyl-inositol-4-phosphate (LPI-4-P), lysophosphatidyl-inositol-5- phosphate (LPI-5-P), lysophosphatidyl-inositol-3 ,4-diphosphate (LPI-3,4-P2), lysophosphatidyl-inositol-3, 5-diphosphate (LPI-3,5-P2), lysophosphatidyl- inositol-4, 5-diphosphate (LPI-4,5-P2), and lysophosphatidyl-inositol-3, 4,5- triphosphate (LPI-3,4,5-P3), and phosphatidyl-inositol (PI), and

lysophosphatidyl-inositol (LPI).

In still other illustrative embodiments, the micelle is comprised of one or more phospholipids selected from the group consisting of PEG-poly (ethylene glycol)-derivatized distearoylphosphatidylethanolamine (PEG-DSPE), PEG- poly(ethylene glycol)-derivatized dioleoylphosphatidylethanolamine (PEG- DOPE), polyethylene glycol)-derivatized ceramides (PEG-CER), hydrogenated soy phosphatidylcholine (HSPC), egg phosphatidylcholine (EPC), phosphatidyl ethanolamine (PE), phosphatidyl glycerol (PG), phosphatidyl inositol (PI), monosialoganglioside, sphingomyelin (SPM), distearoylphosphatidylcholine (DSPC), dimyristoylphosphatidylcholine (DMPC), and

dimyristoylphosphatidylglycerol (DMPG).

In still other embodiments, the micelle comprises one or more PEG- containing phospholipids, for example l,2-dioleoyl-s«-glycero-3- phosphoethanolamine-N-[m ethoxy (poly ethylene glycol)] (ammonium salt) (DOPE-PEG), l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(poly ethylene glycol)] (ammonium salt) (DSPE-PEG), 1,2-distearoyl- s«-glycero-3-phosphoethanolamine-N-[amino(poly ethylene glycol)] (DSPE- PEG-NEh) (DSPE-PEG). In the PEG-containing phospholipid, the PEG group ranges from about 2 to about 250 ethylene glycol units, about 5 to about 100, about 10 to 75, or about 40-50 ethylene glycol units. In certain exemplary embodiments, the PEG-phospholipid is l ,2-dioleoyl-.s//-glycero-3- phosphoethanolamine-N-[m ethoxy (poly ethylene glycol)-2000] (ammonium salt) (DOPE-PEG2000), l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [methoxy(polyethylene glycol)-2000] (ammonium salt) (DSPE-PEG2000), 1,2- distearoyl-s«-glycero-3-phosphoethanolamine-N-[amino(poly ethylene glycol)- 2000] (DSPE-PEG2000-NH2) which can be used to covalent bind a functional moiety to the lipid bi-layer.

Cargo or other functional molecules for inclusion in the single

MOP@micelle or MOP Sa @micelle include but are not limited to“anti-cancer agents” including but not limited to everolimus, trabectedin, abraxane, TLK 286, AV-299, DN-101 , pazopanib, GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY- 142886), AMN-107, TKI-258, GSK461364, AZD 1 152, enzastaurin, vandetanib, ARQ-197, MK-0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, a VEGFR inhibitor, an EGFR TK inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, a Bcl-2 inhibitor, an HD AC inhibitor, a c-MET inhibitor, a PARP inhibitor, a Cdk inhibitor, an EGFR TK inhibitor, an IGFR- TK inhibitor, an anti-EGFR antibody, an anti-HGF antibody, a PI3 kinase inhibitors, an AKT inhibitor, a JAK/STAT inhibitor, a checkpoint-1 or 2 inhibitor, a focal adhesion kinase inhibitor, a Map kinase kinase (mek) inhibitor, a VEGF trap antibody, pemetrexed, erlotinib, dasatanib, nilotinib, decatanib, panitumumab, amrubicin, oregovomab, Lep-etu, nolatrexed, azd2171, batabulin, ofatumumab, zanolimumab, edotecarin, tetrandrine, rubitecan, tesmilifene, oblimersen, ticilimumab, ipilimumab, gossypol, Bio 1 11 , 131-I-TM-601 , ALT- 110, BIO 140, CC 8490, cilengitide, gimatecan, IL13-PE38QQR, INO 1001 , IPdRi KRX-0402, lucanthone, LY 317615, neuradiab, vitespen, Rta 744, Sdx 102, talampanel, atrasentan, Xr 311 , romidepsin, ADS- 100380, sunitinib, 5- fluorouracil, vorinostat, etoposide, gemcitabine, doxorubicin, liposomal doxorubicin, 5'-deoxy-5-fluorouridine, vincristine, temozolomide, ZK -304709, seliciclib; PD0325901 , AZD-6244, capecitabine, L-Glutamic acid, N-[4-[2-(2- amino-4,7-dihydro-4-oxo-lH-pyrrolo[2,3-d]pyrimidin-5-yl)ethy l]benzoyl]-, disodium salt, heptahydrate, camptothecin, PEG-labeled irinotecan, tamoxifen, toremifene citrate, anastrozole, exemestane, letrozole, DES(diethylstilbestrol), estradiol, estrogen, conjugated estrogen, bevacizumab, IMC-1C11, CHIR-258, 3-[5-(methylsulfonylpiperadinemethyl)-indolyl]-quinolone, vatalanib, AG- 013736, AVE-0005, the acetate salt of [D-Ser(But)6,AzglylO] (pyro-Glu-His- Trp-Ser-Tyr-D-Ser(But)-Leu-Arg-Pro-Azgly-NH2 acetate [C59H84N18O14 - (C 2 H 4 0 2 ) X where x = 1 to 2.4], goserelin acetate, leuprolide acetate, triptorelin pamoate, medroxyprogesterone acetate, hydroxyprogesterone caproate, megestrol acetate, raloxifene, bicalutamide, flutamide, nilutamide, megestrol acetate, CP-724714, TAK-165, HKI-272, erlotinib, lapatinib, canertinib, ABX- EGF antibody, erbitux, EKB-569, PKI-166, GW-572016, Ionafamib, BMS- 214662, tipifamib, amifostine, NVP-LAQ824, suberoyl anilide hydroxamic acid, valproic acid, trichostatin A, FK-228, SU11248, sorafenib, KRN951,

aminoglutethimide, amsacrine, anagrelide, L-asparaginase, Bacillus Calmette- Guerin (BCG) vaccine, bleomycin, buserelin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, diethylstilbestrol, epirubicin, fludarabine, fludrocortisone, fluoxymesterone, flutamide, gemcitabine, gleevac, hydroxyurea, idarubicin, ifosfamide, imatinib, leuprolide, levamisole, lomustine, mechlorethamine, melphalan, 6-mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, octreotide, oxaliplatin, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, teniposide, testosterone, thalidomide, thioguanine, thiotepa, tretinoin, vindesine, 13-cis-retinoic acid, phenylalanine mustard, uracil mustard, estramustine, altretamine, floxuridine, 5-deoxyuridine, cytosine arabinoside, 6-mercaptopurine, deoxycoformycin, calcitriol, valrubicin, mithramycin, vinblastine, vinorelbine, topotecan, razoxin, marimastat, COL-3, neovastat, BMS-275291 , squalamine, endostatin, SU5416, SU6668,

EMD121974, interleukin- 12, IM862, angiostatin, vitaxin, droloxifene, idoxifene, spironolactone, finasteride, cimetidine, trastuzumab, denileukin diftitox, gefitinib, bortezomib, paclitaxel, cremophor-free paclitaxel, docetaxel, epithilone B, BMS- 247550, BMS-310705, droloxifene, 4-hydroxytamoxifen, pipendoxifene, ERA- 923, arzoxifene, fulvestrant, acolbifene, lasofoxifene, idoxifene, TSE-424, HMR- 3339, ZK186619, topotecan, PTK787/ZK 222584, VX-745, PD 184352, rapamycin, 40-O-(2-hydroxyethyl)-rapamycin,

temsirolimus, AP-23573, RAD001 , ABT-578, BC-210, LY294002, LY292223, LY292696, LY293684, LY293646, wortmannin, ZM336372, L-779,450, PEG- filgrastim, darbepoetin, erythropoietin, granulocyte colony-stimulating factor, zolendronate, prednisone, cetuximab, granulocyte macrophage colony- stimulating factor, histrelin, pegylated interferon alfa-2a, interferon alfa-2a, pegylated interferon alfa-2b, interferon alfa-2b, azacitidine, PEG-L- asparaginase, lenalidomide, gemtuzumab, hydrocortisone, interleukin- 11, dexrazoxane, alemtuzumab, all-transretinoic acid, ketoconazole, interleukin-2, megestrol, immune globulin, nitrogen mustard, methylprednisolone,

ibritumomab tiuxetan, androgens, decitabine, hexamethylmelamine, bexarotene, tositumomab, arsenic trioxide, cortisone, etidronate, mitotane, cyclosporine, liposomal daunorubicin, Edwina-asparaginase, strontium 89, casopitant, netupitant, an NK-1 receptor antagonists, palonosetron, aprepitant,

diphenhydramine, hydroxyzine, metoclopramide, lorazepam, alprazolam, haloperidol, droperidol, dronabinol, dexamethasone, methylprednisolone, prochlorperazine, granisetron, ondansetron, dolasetron, tropisetron,

pegfilgrastim, erythropoietin, epoetin alfa, darbepoetin alfa and mixtures thereof; anti-HIV agents” including but not limited to nucleoside reverse transcriptase inhibitors (NRTI), other non-nucleoside reverse transcriptase inhibitors (i.e., those which are not representative), protease inhibitors, fusion inhibitors, among others, exemplary compounds of which may include, for example, 3TC

(Lamivudine), AZT (Zidovudine), (-)-FTC, ddl (Didanosine), ddC (zalcitabine), abacavir (ABC), tenofovir (PMPA), D-D4FC (Reverset), D4T (Stavudine), Racivir, L-FddC, L-FD4C, NVP (Nevirapine), DLV (Delavirdine), EFV

(Efavirenz), SQVM (Saquinavir mesylate), RTV (Ritonavir), IDV (Indinavir), SQV (Saquinavir), NFV (Nelfinavir), APV (Amprenavir), LPV (Lopinavir), fusion inhibitors such as T20, among others, fuseon and mixtures thereof.

Other cargos include but are not limited other nanoparticles, such as gold NP and quantum dots.

The micelles or superassemblies may comprise targeting molecules, e.g., a cell targeting species (e.g., a peptide, antibody, such as a monoclonal antibody, an affibody or a small molecule moiety which binds to a cell, among others); a fusogenic peptide that promotes endosomal escape of protocells; a cargo, including one or more drugs (e.g., an anti-cancer agent, anti-viral agent, antibiotic, antifungal agent, etc.); a polynucleotide, such as encapsulated DNA, double stranded linear DNA, a plasmid DNA, small interfering RNA, small hairpin RNA, microRNA, a peptide, polypeptide or protein, an imaging agent, or a mixture thereof, among others), wherein one of said cargo components is optionally conjugated further with a nuclear localization sequence. In certain embodiments, the micelle or lipid may comprise PEG groups and/or targeting peptides. A targeting species including, for example, targeting peptides including oligopeptides, antibodies, aptamers, and PEG (polyethylene glycol) (including PEG covalently linked to specific targeting species); a cell penetration peptide such as a fusogenic peptide or an endosomolytic peptide as otherwise described herein. Targeting peptides may be complexed or covalently linked to the lipid layer through use of a crosslinking agent.

The invention will be further described by the following non

limiting example.

Example 1

Materials and Methods

Reagents. All chemicals and reagents were used as received. 4-Pyridylboronic acid pinacol ester, tetrakis(triphenylphosphine) palladium(O), potassium phosphate, 1,4-dioxane, 2,5-dibromo-3-dodecylthiophene, chloroform, Pd(BF4)2, ethyl acetate, diethyl ether, polyoxyethylene (6) lauryl ether, polyoxyethylene (10) lauryl ether, polyoxyethylene (23) lauryl ether, dimethyl sulfoxide (DMSO), doxorubicin (DOX), sulforhodamine B, Mn(III)tetra (4-sulfonatophenyl) porphyrin, 6-aminocoumarin, fluorescein isothiocyanate, CdSe/ZnS quantum dots (QDs), Ham's F-12K (Kaighn's) medium, Iscove's modified Dulbecco's media (IMDM), and formaldehyde solution (36.5-38% in H2O) were purchased from Sigma- Aldrich. Au NPs were synthesized according to the reported literature. Epidermal growth factor (EGFR)-biotin and NeutrAvidin were purchased from Thermo Fisher Scientific. l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[biotinyl(poly ethylene glycol)-2000] (ammonium salt)) (DSPEPEG-2000-biotin) and l,2-distearoyl-sn-glycero-3-phosphoeth- anolamine-N-[methoxy (polyethyleneglycol)-2000] (ammonium salt) (DSPE- PEG-2000) were purchased from Avanti Polar Lipids. Heat-inactivated fetal bovine serum (FBS), 10x phosphate-buffered saline (PBS), 0.5% trypsin- ethylenediaminetetraacetic acid (EDTA) solution, and penicillin-streptomycin (PS) were purchased from Gibco (Logan, UT). Dulbecco's modification of Eagle's medium (DMEM) was obtained from Coming Cellgro (Manassas, VA), Absolute (200 proof) ethanol was obtained from Pharmco-Aaper (Brookfield, CT). CellTiter-Glo 2.0 assay kit was purchased from Promega (Madison, WI). Hoechst 33342 were obtained from Thermo Fisher Scientific (Rockford, IL). 1 x PBS, Alexa Fluor 488 phalloidin, and rhodamine phalloidin were purchased from Life Technologies (Eugene, OR). Milli-Q water with a resistivity of 18.2 MW cm was obtained from an inline Millipore RiOs/Origin water purification system.

Characterization. The morphology of the samples was characterized by field-emission gun scanning transmission electron microscopy (STEM, JEOL 2010F) at 200 kV and transmission electron microscopy (TEM, Hitachi H-7650) at 200 kV. Argon adsorption-desorption isotherms were obtained using a Quantachrome ASiQ2 intrument at 87 K. ' H NMR spectra were obtained using a JEOLTNM-ECA300 at 300 MHz. Atomic force microscopy (AFM) images were acquired using an Asylum Research MFP-3D™ AFM. UV-Vis absorption spectra were recorded using a Perkin-Elmer UV/vis Lambda 35 spectrometer. The fluorescence emission measurements were carried out using a fluorescence spectrometer (Perkin-Elmer LS55). Fluorescence images were acquired using a Zeiss LSM510 META (Carl Zeiss Microimaging, Inc.; Thomwood, NY, USA) operated in channel mode of the LSM510 software. The software used for the optimization of the structure of Pd 24 L 48 -Ci 2 MOP was Materials Studio 8.0. Due to the large coordination structure of Pd 24 L 48 -Ci 2 MOP (more 3,000 atoms), only molecular mechanics (MM) simulation was used.

Ligand 1 synthesis

For the synthesis of ligand 1, 2.10 g 4-pyridylboronic acid pinacol ester (9.80 mmol), 0.407 g tetrakis(triphenyl- phosphine)palladium(O) (0.352 mmol), and 5.95 g potassium phosphate (28.0 mmol) were first added into a three-neck flask. Under the protection of argon atmosphere, 70 mL 1,4-dioxane and 1.121 mL 2,5-dibromo-3-dodecylthiophene (3.61 mmol) were then added, and the suspension was stirred at 90°C for 3 days. After cooling down, the residue was filtered and washed with chloroform. The filtrate was further purified by silica gel column chromatography to give a yellow solid.

Pd24L48-Ci2 MOP synthesis For the preparation of PCI24L48-C12, 31.0 mg ligand 1 (76.4 pmol) was mixed with 8.80 mg Pd(BF4)2 (38.2 pmol) in a mixture of acetonitrile and chloroform (3 :1 v/v) and reacted at 70°C for 24 hours. After cooling down, an excess amount of a mixture of ethyl acetate and diethyl ether (1 : 1 v/v) was added to the solution to promote precipitation. The precipitate was centrifuged (10,000 rpm, 10 minutes) and dried in vacuum to give the desired cage as a light yellow solid.

MOP super-assembly

For the synthesis of single MOP@micelle, 10 mg mL-1 of

polyoxyethylene (6) lauryl ether-based micelle was first prepared. Then, a small drop of MOP solution (0.038 mM in DMSO) was added into the micelle solution followed by sonication to promote encapsulation. For the synthesis of

MOP sa @micelle with different sizes, 1 mg mL 1 of polyoxyethylene (6) lauryl ether-based micelle was prepared and then different amounts of Pd 24 L 48 -Ci 2 MOP (0.17 mM in DMSO) were added, followed by sonication to promote super-assembly. To vary the length of PEG outside the micelle, an alternative assembly unit, e.g., polyoxyethylene (10) lauryl ether or polyoxyethylene (23) lauryl ether, was used.

Guest loading number calculation

The loading number of guest molecules inside Pd 24 L 48 -Ci 2 MOP was determined by UV-vis spectroscopy. The fitting of the absorbance versus the concentration of the guest molecules and Pd 24 L 4 x-C 12 MOP was first carried out. The characteristic absorption wavelengths of Pd24L48-Ci2 MOP and the guest molecules (DOX, sulforhodamine B, and Mn(III)tetra (4-sulfonatophenyl) porphyrin) were found to be 365, 503, 561, and 467 nm, respectively. As shown in Figure 3a and Figure 12, the MOP of Pd24L48-Ci2 displayed no absorption at the characteristic absorption peak positions observed for the guest molecules. From the UV-vis spectra of the guest molecule-loaded MOP sa @rnicelles, the concentration of various guest molecules can be determined. After subtraction of the absorption at 365 nm, originating from the guest molecules, the residual absorption at 365 nm that is attributed to the adsorption of Pd24L48-Ci2 cage can be used further for cage concentration fitting. Based on the above couple of steps, the loading number of the guest molecules inside Pd 24 L 48 -Ci 2 MOP can be determined. Separation membrane fabrication

A microtube that contains porous polypropylene membrane was used.

100 pL Pd 24 l- 48 -Ci 2 MOP@micelle (1 mg mL 1 ) solution was added into the microtube and then centrifuged at 10,000 rpm for 10 minutes. This coating process was repeated for a couple of times until the desired thickness was achieved. During each coating, the membrane was washed by water once.

Hemolysis assay

The purified red blood cells (RBCs) were incubated with different concentrations of particles at room temperature for 3 h in continuous rotating state. Double distilled (DI) water and lx PBS containing purified RBCs were used as the positive and negative controls, respectively. Finally, the mixtures were centrifuged at 300 g for 3 minutes, and 100 pL supernatant of all samples was transferred to a 96-well plate. The absorbance of hemoglobin in the supernatant was measured by a BioTek microplate reader (Winooski, VT) at 540 nm. The hemolysis percentage of each sample was determined using the reported equation. 2 Percent hemolysis (%) = 100 c (Sample Abss4o mn - Negative control Abs54o mn)/(Positive control Abss4o mn /Negative control Abss4o mn).

Drug release

For the drug release studies, 2 mg of DOX-loaded MOP Sa @micelle in 1.2 mL PBS buffer solution (pH 7.4 or 5.5) was loaded into a small tube at room temperature. During each time interval, the nanoparticles were centrifuged (20,000 rpm, 10 minutes), and half of the supernatant solution was withdrawn, followed by the addition of 0.6 mL fresh PBS buffer. The content of DOX was determined by UV-vis titration.

Cell culture

Cell culture was performed using standard procedures (atcc.org). For adherent cells, HeLa (CCL-2) and A549 (CCL-185) were obtained from

American Type Culture Collection (ATCC) and respectively stored in DMEM and F-12K media containing 10% FBS at 37°C and 5% CO2. Cells were passaged at approximately 80% confluency. For coating purposes, living adherent cells (HeLa and A549) were removed from plate bottom using Trypsin- EDTA (0.25%) and then suspended in culture media.

Cell viability testing Cell viability of the constructed nanocarriers was assessed by CellTiter- Glo 2.0 assay. Briefly, single-MOP@micelle or MOP Sa @micelle nanocarriers were first diluted to a concentration of 50000 cells mL 1 . Then, 100 pL of the samples was added into a white 96-well plate. Subsequently, 100 pL CellTiter- Glo 2.0 reagent was dispensed into each well. The luminescence was recorded 10 minutes after addition of CellTiter-Glo 2.0 reagent by a BioTek microplate reader. Cell viability was calculated as a percentage of mammalian cells in the absence of nanocarriers.

Anti-EGFR modification

First, polyoxyethylene (23) lauryl ether, DSPE-PEG-biotin, and DSPE- PEG were mixed at mol% ratio of 92:4:4, and then dried under high vacuum to remove the organic solvent. Then, the dried film was hydrated in lx PBS, and the bath was sonicated for 30 minutes to obtain a micelle solution at a concentration of 1 mg

mL 1 . Then, 150 pL fluorescent dye labeled Pd24L48-Ci2 MOP in DMSO was added to the micelle solution, followed by sonication to promote MOP super assembly. The obtained MOP sa @micelle-based nanocarriers were washed with lx PBS twice. For anti-EGFR modification, 200 pL neutravidin protein (3 mg mL 1 ) was added to the nanocarriers. After incubation for 30 minutes, the particles were centrifuged (20,000 rpm, 10 minutes), and the supernatant was removed. After redispersion in lx PBS, 200 pL biotin-EGFR (0.2 mg mL 1 ) was added and incubated at room temperature for 30 minutes. After washing with 1 c PBS twice and redispersion in 100 pL PBS, the antibody-conjugated

nanocarriers can be directly used for in vitro targeting experiments.

In vitro targeting

For the studies, 2 c 10 5 A549 (CCL-185, ATCC) cells in 6-well plates with 2 mL F-12K media containing 10% FBS and 1% PS were seeded and then incubated at 37 °C in 5% C0 2 -humidified atmosphere. After 24 hours, the media was removed and replaced with 1 mL fresh complete cell culture media supplemented with 50 pg mL 1 of EGFR-modified or unmodified

MOP sa @micelles for different times at 37°C under 5% CCh-humidified atmosphere. After incubation, the media was removed, and the cells were gently washed twice with lx PBS. For imaging purposes, the treated cells were fixed in 4% paraformaldehyde (in lx PBS) at room temperature for 15 minutes, washed with lx PBS twice, and then stored in 1 mL lx PBS. The cell nuclei and F-actin were stained with 1 mL Hoechst 33342 (3.2 mM lx PBS) for 30 minutes and 200 pL Alexa Fluor488 phalloidin (20 nM in lx PBS) for 45 minutes, respectively. After staining, the cells were washed with lx PBS twice and stored in lx PBS prior to fluorescence microscope imaging.

Pharmacokinetics and biodistribution studies

All the animal procedures complied with the guidelines of the University of New Mexico Institutional Animal Care and Use Committee and were conducted following institutional approval (Protocol 17-200658-HSC). The experiments were performed on female Albino C57BL/6 mice (6 weeks). To evaluate the circulation half-life, 150 pL of CdSe/ZnS QD (627 nm)-labeled MOP sa @ micelles (1 mg/mL) were injected into the eye of the mice. The blood was collected at 0.5, 1, 2, 6, 12, 24, and 48 hours following the injection. Each time point group contained three mice. The collected blood samples were diluted with the same amount of IX PBS before fluorescence measurement. Particle retention in circulation at these time points was determined by measuring the fluorescence on a BioTek microplate reader (Winooski, VT). Pharmacokinetics parameters were calculated to fit a non-compartment model.

To study the related biodistribution in various tissues, 150 pL of

CdSe/ZnS QD (627 nm)-labeled MOP sa @ micelles (1 mg/mL) were retro-orbital injected to mice. At each of the 6, 12, 24, and 48 hours time points following the NP injection, three mice were randomly selected and euthanized. Their spleen, liver, heart, lung, and kidneys were collected. The collected organs were examined with an IVIS fluorescence imaging system (Xenogen, Alameda, CA), and the fluorescence intensity of the MOP particles in different organs was further semi-quantified by the IVIS imaging software.

Results

The superassembly of MOP is described herein. The fabrication technique affords a simple synthesis process and the organization of individual MOP to form advanced hierarchical structures. Specifically, the superassembly of MOP supported micelles (MOP Sa @micelle) for targeted drug delivery is described (see Scheme la in Figure 1). The key fabrication point is the decoration of the outer surface of the MOP with hydrophobic chains to transform MOP units into nanobuilding blocks that can self-assemble into larger and well- defined superassemblies within micelles. This approach is different from the reported molecular building block (MBB) approach that is used to construct metal-organic framework (MOF)-based lipid bilayers (Zhu et ah, 2018), where the open metal sites of MOF units allow further coordination of additional organic ligands to form MOF NPs (Scheme lb in Figure 1). Nonetheless, the MBB approach typically affords limited size control, whereas the superassembly approach described herein provides homogeneous size distributions in broad ranges of diameters through the simple control of the MOP concentration to obtain ultrasmall single MOP@micelle and MOP sa @micelle. In addition, owing to the highly porous nature of the MOP, various cancer drugs were controllably loaded into individual MOP units with high payloads and subsequently integrated into MOP superassemblies to form multiple drug delivery systems. Furthermore, the MOP superassembly approach enabled the modular assembly of MOPs with additional functional NPs such as fluorescent quantum dots (QDs) for multifluorescence imaging or gold NPs for enhanced bioimaging. To demonstrate the potential of the MOP superassembly concept for drug delivery, a Fujita-type MOP composed of dozens of dodecyl chains was chosen as a prototype. A dose-dependent assay for MOP sa @micelles of different sizes demonstrated negligible hemolytic activities and long-term colloidal stability in various media, thereby mitigating possible concerns of structure disassembly during circulation. Cell viability tests further demonstrated the good

biocompatibility of the MOP sa @micelles that is related to the NP size and the molecular weight of the polyethylene glycol (PEG) that was used for surface modification. Importantly, designing MOP sa @micelle nanocarriers with targeting moieties enabled targeted cancer cell delivery and in vivo experiments using a mouse model also confirmed the good circulation. Taken together, this novel and modular superassembly approach combines the synergistic advantages of micelles (e.g., low inherent toxicity and long circulation time) and the MOP superassembly (e.g., highly controlled architecture, stability, and high payloads of multiple cargos), and promotes the design of MOP -inspired nanocarriers for targeted cancer therapies.

A giant Pd24L48 Fujita-type spherical framework (~7.2 nm) composed of up to 48 decorations of dodecyl chains (denoted as Pd24L4x-C 12) was first prepared for proof of concept (Figure la) (Sun et al, 2016). Dipyridyl-3- dodecyl-thiophene was first synthesized as an organic linker (ligand 1) using the Suzuki-Miyaura reaction. To prepare the coordination sphere, ligand 1 (0.1 mmol) and Pd(BF4)2 (50 pmol) were heated in a mixture of acetonitrile and chloroform at 70°C for 24 hours. Then, an excess amount of a mixed solution of ethyl acetate and diethyl ether (1 :1 v/v) was added to promote the precipitation. The structure of Pd24L48-Ci2 MOP was first determined by Fourier-transform infrared spectroscopy. As shown in Figure 6, the characteristic peak at 1589 cnT 1 assigned to the vibration of C=N in the pyridine ring was shifted to 1610 cm , indicating the coordination of Pd 2+ metal ions. Based on chemical analysis, the ratio of organic ligand to Pd 2+ was calculated to be ~1.8, which is close to the exact ratio of 2. Moreover, in nuclear magnetic resonance (NMR) spectra (Figure IB), the b and c protons of the pyridyl or thiophene groups (¾ and H c ) were shifted downfield by 0.20 and 0.22 ppm, respectively, upon coordination to Pd 2+ . Compared with the ¾ signal of free ligand 1, the ¾ signal of Pd 24 l- 48 -Ci 2 MOP was much broader. Diffusion-ordered NMR measurements also

demonstrated a single product given the diffusion coefficient of 5.7 c 10 10 m 2 s _1 , which was indicative of the formation of larger chemical species.

Furthermore, electrospray ionization and matrix-assisted laser desorption ionization time of flight mass spectrometry techniques were used to confirm the coordination structure. Although the molecular weight of the Pd24l-48 cage was determined by a series of prominent peaks in (m!z = 17, 24, 27, and 33; Figure 7), the molecular weight of the Pd 24 L 48 -Ci 2 cage could not be quantified by the same technique, presumably due to presence of the multiple dodecyl chains. Nevertheless, high-resolution transmission electron microscopy (HR-TEM) displayed a dense packing of Pd24L48-Ci2 MOPs with a distance of 4.8 nm between closest neighbors (Figure 1C). Atomic force microscopy (AFM) imaging also showed single cages with sizes of -5.0 nm (Figure 8). All this structural information confirmed the successful formation of giant coordination spheres. Based on molecular mechanics (MM) calculations, the optimized structure of Pd24L48-Ci2 was obtained. As shown in Figure ID, the

rhombicuboctahedral structure of Pd 24 l- 48 -Ci 2 is highly spherical with an inscribed sphere with a diameter of 3.6 nm and a circumscribed sphere, with alkane chains, of 7.2 nm in diameter. The distance between antipodal palladium atoms was measured to be 4.0 nm.

Subsequently, the preparation of single MOP@micelle NPs followed by MOP superassembly was performed using the Pd 24 l- 48 -Ci 2 MOP nanobuilding blocks. First, to promote the superassembly of the hydrophobic Pd 24 L 48 -Ci 2 MOP cages in an aqueous solution, a micellar solution assembled by

polyoxyethylene (6) lauryl ether (Pr,LEL) was added (Figures 1A,E,F). A small drop (1 pL) of an MOP solution (0.17 x 10 3 m in dimethyl sulfoxide (DMSO)) was added to the micellar solution, and the resulting mixture was sonicated to promote the formation of single MOP-supported micelle NPs (Figure 2A). High- magnification dark-field scanning transmission electron microscopy (STEM) of the superassembly showed uniform sizes of 4.2 ± 0.7 nm (Figure IE), well correlated with the molecular shell of Pd 24 L 48 -Ci 2 MOP defined by the coordinated palladium ions. The hydrodynamic diameters of the micelle and MOP-micelle conjugate were determined by dynamic light scattering (DLS) to be 13.6 and 18.8 nm (Figure IF), respectively. These data confirm the successful formation of single MOP-supported micelles. Moreover, with the increase of MOP concentration, larger MOP superassemblies with nanometer or micrometer sizes were generated (Figures 2A,B). Notably, the size could easily be tuned in a broad range by gradually increasing the MOP concentration (Figure 2C). The monodisperse particle size distributions of the single MOP-supported micelle (6.8 ± 0.6 nm) and MOP superassemblies (65.8 ± 4.8 and 100.4 ± 14.6 nm) were further confirmed by AFM images (Figure 2D; Figure 8). TEM imaging of the air-dried samples revealed a hexagonal packing of MOPs inside the

superassemblies with a long-range order on a scale of 100 nm x 100 nm (Figure 2E). The enlarged image shown in Figure 2F reveals the overlaying hexagons with a 5.4 nm dimension. The hydrophobic interactions from the long alkane chains of closest MOPs are presumably the main driving force for the superassembly of MOP and may as a result lead to high stability during in vivo circulation. The surface area and pore volume of the MOP sa @micelle NPs were found to be inaccessible using standard argon adsorption-desorption

measurements due to the kinetically closed pore of the cages and shell blocking effect as a result of dense packing of the long PEG chains (Figure 10) (Park et al, 2014). The cargo loading capabilities of the MOP Sa @micelle nanomaterials were then investigated. The high inherent porosity of Pd24L48-Ci2 as well as the relatively large pore windows (1.4 nm x 1.4 nm) of the cages (Figure 11) suggested that various guest molecules could be loaded into the MOP nanocavity in a controlled manner. To evaluate the cargo loading capacities of our nanocarriers, three types of molecules were separately loaded, including doxorubicin hydrochloride (DOXTTC1) as a hydrophilic drug for chemotherapy, sulforhodamine B as a fluorescent dye for labeling, and Mn(III)tetra (4- sulfonatophenyl) porphyrin (MnTPPS4) as a contrast agent for magnetic resonance imaging. The successful loading of the guest molecules was confirmed in the particles after cargo loading by the characteristic absorption bands of the guest molecules displayed in the UV-vis spectra (see 503, 561, and 467 nm in Figure 3A and Figure 12). Based on calculations from the UV-vis spectra, one Pd 24 l- 48 -Ci 2 MOP entity was estimated to be loaded with 22 DOXTTCl, 38 sulforhodamine B, and 13 MnTPPS4, confirming the high loading efficacy. Note that the structure of the cargo-loaded MOP Sa @micelle

nanocarriers remained unaffected by the cargo loading (see TEM image in Figure 13). In addition, multiple guest molecules were loaded within individual MOP, which were then superassembled simultaneously with a highly controlled molar ratio. For instance, three fluorescent molecules (6-aminocoumarin, fluorescein isothiocyanate, and sulforhodamine B) were individually loaded into different Pd 24 L 48 -Ci 2 MOP samples and then integrated into MOP

superassemblies with tunable sample ratios for potential multifluorescence imaging, as confirmed by UV-vis spectroscopy (Figure 3B). This controllable superassembly scheme provides a powerful approach to prepare multidrug delivery nanosystems with precise control of the ratio for each drug by simply tuning the MOP sample ratios.

The superassembled MOP-micelle particles were then incorporated with functional materials and nanomaterials for a range of proof-of-concept studies. First, several functional nanomaterials, such as fluorescent QDs and gold nanomaterials (Figure 14), were incorporated into the MOP Sa @micelle carriers via one-pot during the superassembly of MOP-micelle nanocarriers. As depicted in Figure 3C, the fluorescent peak at 627 nm for CdSe/ZnS quantum dot@ MOP sa @micelle and the typical plasmon resonance peak at 529 nm for Au NPs@ MOPsa@micelle confirmed the successful doping of the MOP Sa @micelle particles (Figures 16-17). Such incorporation of functional nanomaterials into the MOP sa @micelle nanocarriers can diversify the utility and potential of the platform. Additionally, it was found that the MOP superassemblies could be deposited onto polypropylene membranes as prototypes for separation applications (Figure 3D). The fabrication of the mixed membrane was realized with a controlled thickness of ~2 pm, as shown by SEM (Figure 3E). After five deposition cycles (Figure 3D), a film consisting of a dense packing of MOP superassemblies was obtained and was then applied to separate mixtures. Owing to the porosity and the reversible host-guest interactions of MOPs, the resultant membrane showed promising separation properties for sulforhodamine B- containing solutions, even after a few cycles of separation (Figure 3F). All of these results point to multifunctional MOP Sa @nricelle nanocarriers and novel mixed membranes based on MOP nanocages for biomedical and separation applications.

The biocompatibility and colloidal stability of the MOP sa @ micelles were then studied in view of future biomedical applications. To evaluate the concentration-dependent lysis of red blood cells (RBCs), the nanomaterials were incubated with RBCs at concentrations varying from 1 to 200 pg mL 1 in l x phosphate-buffered saline (PBS) solution for 3 hours. As shown in Figures 4A- B, the hemolysis percentage of RBCs for all samples increased in a dose- dependent manner. The smaller single MOP@micelle carriers caused a higher release of hemoglobin than the larger carriers. The same trend was observed with mesoporous silica nanoparticles (Lin et al, 2010). Nevertheless, all of the nanocarriers showed negligible hemolytic activities, thus meeting the essential prerequisites of biomedical applications. The colloidal stabilities of the MOP sa @ micelles were then tested in various media (PBS solution, Dulbecco’s modified Eagle medium (DMEM), and F-12K). DLS data of the MOP sa @micelle samples with different sizes showed narrow hydrodynamic size distributions with low PDI values of less than 0.2 (Figure 4C). Notably, long-term exposure in various media (7 days) caused insignificant size changes, indicating long-term colloidal stability. It can be surmised that the polymeric PEG chains act as protective layers to prevent particle aggregation. The cytotoxicity profiles of the single MOP@micelle or MOP Sa @rnicelle-based nanocarriers with different PEG chain lengths and sizes were also evaluated against two human cancer cell lines: A549 cell (lung cancer cell) and HeLa cell (cervical cancer cell). The corresponding ICso values (concentration required to reduce cell viability to 80%) are summarized in Figure 4e. As shown in Figure 4E and Figure 17, free nanometric MOP exhibited higher cytotoxicity (ICso = 25 pg mL ') on A549 cells than MOP superassemblies with different PEG chain lengths (6, 10, and 23) (ICso = 62,

103, and 200 pg mL 1 , respectively), thus confirming the low inherent toxicity of PEGylation. In addition, higher PEG repetitive unit numbers ranging from 6 to 23 led to considerably reduced cytotoxicity for the MOP superassemblies, which correlates with the reported molecular weight-dependent cytotoxicity of PEG samples (Pozzi et al., 2014). Additionally, size-dependent cytotoxicity effects were observed (Figure 18). For ultrasmall single MOP@micelle particles, the ICso value (82 pg mL 1 ) was much higher than that for MOP sa @ micelles with larger sizes. In summary, increasing the particle size leads to reduced toxicity, and the high cell viabilities of all MOP@micelle-based nanocarriers (even at high concentrations) suggest the good biocompatibility of the hybrid

nanocarriers developed herein for biomedical applications.

The potential of the biocompatible MOP-micelle carriers for cargo release was then tested in aqueous solution. The DOX drug was selected as it is a widely used anticancer drug. Drug release experiments were carried at room temperature in fresh PBS buffer solution at pH 7.4 and 5.5. The absorbances of the supernatant collected at fixed time intervals were measured to determine the amount of DOX released under these conditions. As displayed in the release profiles of Figure 4D, nearly 36% of the total drug release at pH 7.4 and 5.5 occurred within the first 4 hours, and 5% more of the total drug was released after 7 hours. In this configuration of MOP and cargo, the pH had negligible influence on the release kinetics. Note that DOX may have quite strong host- guest interactions with Pd 24 L 48 -Ci 2 MOP due to collective, weak, noncovalent interactions. When the concentration of DOX inside the MOP cavity was high, the interactions between DOX and MOP may be weakened to a certain extent due to both DOX/DOX and DOX/MOP interactions. Following the release of DOX from the MOP cavity, there may be a balance between releasing and rebinding, which leads to only a partial release of loaded drugs. Nevertheless, given the versatility of MOP and drug structure, controllable release via stimuli - responsive MOPs (e.g., light) could be envisioned to promote the construction of smart drug delivery systems with higher extents of release (Han et al, 2013).

The potential of the MOP sa @micelle carriers for targeted drug delivery was eventually assessed on cancer cells. The A549 cancer cell line was selected owing to the high expression of the epidermal growth factor receptor (EGFR). Targeting was accomplished through the conjugation of biotin-NeutrAvidin- biotin moieties onto MOP Sa @micelle with anti-EGFR monoclonal antibodies as depicted in Figure 5 A. The preparation involved polyoxyethylene (23) lauryl ether (P23LEL): l,2-distearoyl-sn-glycero-3-phosphoethanolamine-A f - [biotinyl(poly ethylene glycol)-2000] (ammonium salt)) (DSPE-PEG-2000- biotin): l,2-distearoyl-.s//-glycero-3-phosphoeth-anolamine-A f - [methoxy(polyethyleneglycol)-2000] (ammonium salt) (DSPE-PEG2000) with molar ratios of 92:4:4. To examine the targeting specificity, MOP sa @micelles with or without EGFR modification were incubated with A549 cells. The confocal laser scanning microscopy images of A549 cells cultured with both red fluorescent dye sulforhodamine B-labeled MOP sa @micelles (50 pg mL at 37°C for 45 minutes are shown in Figures 5B-C. The cellular filamentous actin network and nuclei were stained with fluorescent probes Alexa Fluor 488 phalloidin and Hoechst 33 342, respectively. Significant selective binding and internalization of the EGFR-modified MOP sa @micelles to A549 cells were observed. Binding or internalization of particles without EGFR functionalization was not observed. It was thus concluded that the MOP sa @micelles were mostly internalized into the cells via receptor-mediated endocytosis. To elucidate the receptor-ligand binding kinetics, both dye-labeled MOP sa @ micelles were incubated with A549 cells for different times. After incubation for 15 minutes, a small binding shift occurred for the control samples, even after extending the incubation time to 45 minutes (Figures 5D,E). Regarding the EGFR-modified samples, however, an obvious binding shift was observed within 15 minutes, and a maximal binding shift was observed after 45 minutes. The targeted cell killing efficacy of MOP sa @micelles nanocarriers against A549 cells was also measured. The viabilities of the A549 cells after being incubated for 2 hours with a series of concentrations of free DOX and MOP sa @micelles with or without DOX loading were quantified. As shown in Figure 19, free DOX exhibited dose- dependent toxicity to A549 cells. In contrast, MOP Sa @micelles with the same DOX loading displayed a lower toxicity, indicating that DOX had been trapped in the MOP Sa @micelle nanocarrier and thus reduced the killing efficacy of DOX to the cells. To evaluate the target-specific drug delivery, A549 cells were incubated with increasing concentrations of MOP Sa @micelles with or without EGFR modification or DOX loading in complete media under standard culturing conditions. In Figure 5F, compared to the MOP Sa @micelles with (18.0%) or without EGFR modification (0.1%), the DOX-loaded targeted MOP Sa @micelle causes much higher cell death of -42.0%, indicating an enhanced killing efficacy that was attributed to the successful release of DOX inside A549 cells.

Furthermore, to evaluate the potential in vivo application of the MOP sa @ micelles, the related circulation and biodistribution were assessed using a mouse model. Mice were injected with CdSe/ZnS QD (627 nm) labeled MOP sa @ micelles by retro-orbital injection at a dose of 150 pg NPs/mouse. To study the circulation half-life, at various time points following the injection, blood was collected from the eye socket of the mice to evaluate the NPs remaining in circulation. At 24 and 48 hours postinjection, the MOP sa @ micelles exhibited 12% and 3% overall retention in mice blood (Figure 19), respectively, similar to the 11% and 2% exhibited by PEG-coated NPs (Hu et al, 201 1). The semilog plot of retention-circulation time (Figure 21) illustrates an exponential decrease in particle concentration over time, indicating that the MOP sa @micelle NP circulation followed a one-way nonlinear clearance model. Based on this pharmacokinetic model, the half-life (i.e., time at which 50% of the particles are cleared) of MOP sa @micelle NP was calculated to be 5.6 hours. Moreover, to analyze the related biodistribution, at 6, 12, 24, and 48 hours postinjection, mice were euthanized and their liver, spleen, kidneys, heart, lungs, and blood were harvested for fluorescence analysis (Figure S16, Supporting Information). The majority of fluorescence signal was found in the two primary filtering organs (Figure 21), the liver and spleen at 6 hours postinjection, supporting removal by the reticuloendothelial system (RES) (Zhang et al, 2009; Cui et al, 2015). This behavior is consistent with the behavior of nanocarriers employed currently for in vivo delivery (Lipka et al, 2010). After 24 hours a visible signal was found in the feces (Figure 22), indicating the MOP sa @micelle NPs can be cleared from the body. These results demonstrate that our developed MOP sa @micelle NP has good in vivo residence time and clearance behavior needed for targeted cancer therapies.

In summary, the superassembly of nanosized metal-organic polyhedra and their biomimetic coatings of lipid bilayers are described herein, which synergistically combine the advantages of micelles and supramolecular coordination cages for biomedical applications. The hydrophobic self-assembly of the MOP nanocages was facilitated via their surface modification using dodecyl groups, and remarkable long-range hexagonal order was visualized via high-resolution electron micrographs. Homogeneous and tunable size distributions of MOP sa @ micelle particles were obtained by simply adjusting the MOP concentration. PEGylated nanomaterials showed good biocompatibility and were stable in biorelevant media, as demonstrated by systematic in vitro studies. The MOP superassembly could be loaded with various dyes and drugs, QDs and gold nanoparticles, and could function as multidrug delivery systems with specific cargo loading amounts through the precise mixing of multiple cargo-loaded MOP samples. Through the surface modification with targeting moieties, the MOP sa @ micelles were successfully synthesized and applied to cancer cells where we observed selective cytotoxicity. Finally, in vivo experiments in a mouse model demonstrated good in vivo circulation of

MOP sa @micelles. The metal-organic polyhedral supra-assembly within micelles concept has thus been validated and expected to be applicable to a range of biomedical applications as well as for separation, sensing, and catalytic processes.

References

Ahmad et al, Chem Soc. Rev.. 44:9 (2015);

Blanco et al., Nat. BiotechnoL 33 :941(2015).

Cook et al, Acc. Chem Res.. 46:2464 (2013).

Croissant et al, Adv. Healthcare Mater.. 7:1700831 (2018).

Croissant et al, Adv. Mater.. 29:1604634 (2017).

Cui et al. ACS Nano 9:1571 (2015).

Elsabahy et al., Chem Rev., 115 :10967 (2015).

Fujita et al, Chem 1:91 (2016).

Grim et al., Chem Soc. Rev., 45 :6520 (2016). Grishagin et al, Proc. Natl. Acad. Sci. USA, 111 :18448 (2014).

Han et al.. Angew. Chem. 125. 1358 (2013).

Han et al., Angew. Chem.. Int. Ed. Engl.. 52:1319 (2013);

Harris et al, Chem. Commun., 49:6703 (2013).

Hu et al., Proc. Natl. Acad. Sci. USA, 108:10980 (2011).

Lin et al., J. Am Chem Soc., 132:4834 (2010).

Lin et al., J. Am Chem Soc., 132:4834 (2010).

Lipka, Biomaterials, 3 6574 (2010).

Mitragotri et al., Nat. Rev. Drug Discovery, 13 :655(2014).

Park et al., Angew. Chem , 126:5952 (2014).

Park et al., Angew. Chem Int. Ed. Engl 53/5842 (2014).

Pozzi et al., Nanoscale. 6:2782 (2014).

Richardson et al., Chem. Rev. 116: 14828 (2016).

Rodriguez et al, J Am Chem Soc. 139:55 (2017).

Samanta et al, J Am Chem Soc.. 138:14488 (2016).

Samanta et al, J Am Chem Soc. 139:9066 (2017).

Schmitt et al., J. Am Chem Soc. 134:754 (2012).

Sun et al, Science, 328:1144 (2010).

Therrien et al, Angew. Chem , 120:3833 (2008).

Therrien et al., Angew. Chem , Int. Ed. Engl., 47:3773 (2008).

Vardhan et al, Coord. Chem Rev., 306: 171 (2016).

Xiong et al. Nano Lett., 13 : 1041 (2013).

Xu et al., Chem - Eur J„ 23:3542 (2017).

Yu et al, Proc. Natl. Acad. Sci. USA, 113 :13720 (2016).

Zhang et al., Biomaterials, 30: 1928 (2009).

Zhao et al, Adv Mater , 23:90 (2011).

Zheng et al., Chem. Sci., 6:1189 (2015).

Zhu et al, Adv. Funct. Mater. 28: 1705274 (2018).

All publications, patents and patent applications are incorporated herein by reference. While in the foregoing specification this invention has been described in relation to certain preferred embodiments thereof, and many details have been set forth for purposes of illustration, it will be apparent to those skilled in the art that the invention is susceptible to additional embodiments and that certain of the details described herein may be varied considerably without departing from the basic principles of the invention.