ENGINEERED CRY PROTEINS FOR DELIVERY OF THERAPEUTICS

Information

  • Patent Application
  • 20230113102
  • Publication Number
    20230113102
  • Date Filed
    April 15, 2020
    4 years ago
  • Date Published
    April 13, 2023
    a year ago
Abstract
Provided are novel recombinant proteins that are capable of self-crystallization and exhibit desirable physical properties such as enhanced cellular uptake or and endoso-mal escape. Polynucleotides encoding the recombinant proteins as well as methods of making and using such proteins are also described.
Description
BACKGROUND OF THE INVENTION

In the past few decades, discoveries about the causes of various human diseases at molecular and cellular levels, combined with technical advances in genetic engineering and pharmaceutical sciences, have enabled treatment of many life-threatening illnesses by administering therapeutic proteins to patients. Depending on the target disease as well as the nature and mechanism of action of the therapeutic proteins, the delivery of therapeutic proteins to a targeted tissue or organ site varies dramatically in its specific routes, protein stability or bioavailability, and therefore its ultimate effectiveness, despite significant effort having been devoted to such research endeavors. To this date, effective delivery remains the major obstacle to achieve desired therapeutic outcome by administration of therapeutic proteins. Thus, there exists an urgent need for developing new and improved strategies for delivering therapeutic proteins for the purpose of medical treatment. This invention addresses this and other related needs.


BRIEF SUMMARY OF THE INVENTION

This invention provides a novel approach to achieve effective delivery of proteins of interest, e.g., therapeutic proteins. Thus, in a first aspect, this invention provides a modified Cry protein such as a polypeptide comprising the amino acid sequence shown in a modified SEQ ID NO:1, with two or more, three, four, five, six, seven, eight, nine, ten or more, or all of amino acids at residues 391, 395, 423, 430, 432, 433, 436, 461, 462, 463, and 466 of SEQ ID NO:1 replaced with charged residues such as lysine or arginine, and the polypeptide forms crystal upon being expressed in a host cell. Optionally, at least one, two, or three, amino acids at residues 533, 535, and 536 of SEQ ID NO:1 are further modified, for instance, substituted with alanine. Additionally, The polypeptide may comprise any three-domain Cry protein with a similar structure to Cry3Aa (SEQ ID NO:1), with two or more, three, four, five, six, seven, eight, nine, ten or more, or all of aspartic acid and glutamic acid residues in the region corresponding to the domain II of Cry3Aa replaced with lysine or arginine, and the polypeptide forms crystal upon being expressed in a host cells. This invention provides additional recombinant polypeptides for the same or similar uses by modifying other Cry proteins in the same or similar manner desribed above and herein. The aspartate and glutamate residues in other Cry proteins that can be modified (e.g., substituted with lysine or arginine) to achieve a similar charge profile are identified as follows:

  • Cry1Aa: residues 308, 332, 379, 384, 387, 409, 412, 418, 460 in SEQ ID NO:4;
  • Cry2Aa: residues 379, 383, 385, 396, 420, 436, 437, 451, 479 in SEQ ID NO:5;
  • Cry4Aa: residues 392, 408, 430, 436, 444, 455, 457, 468, 483, 495 in SEQ ID NO:6;
  • Cry5B: residues 375, 389, 397, 402, 425, 430, 433, 437, 473, 490 in SEQ ID NO:7;
  • Cry7Ca1: residues 389, 394, 430, 431, 451, 456, 459, 460, 465, 467, 468, 478, 482 in SEQ ID NO:8;
  • Cry8Ea1: 403, 430, 435, 449, 455, 457, 459, 461, 465, 468, 473, 489 in SEQ ID NO:9;
  • Cry10Aa: residues 396, 410, 413, 419, 422, 426, 435, 446, 453, 465, 466, 477, 486 in SEQ ID NO:10; and
  • Cry11Aa: residues 381, 422, 443, 455, 457, 460, 469, 473, 484, 487, 498 in SEQ ID NO:11.


In addition, the polypeptide of this invention further includes a fragment of a modified Cry protein (e.g., a modified Cry3Aa protein) generally corresponding to its domain II, such as the 389-471 fragment of SEQ ID NO:2, which the present inventors have discovered to be a soluble peptide and act in a manner similar to that of a cell-penetrating peptide and therefore possesses the capability of effectively transporting its fusion partner, a protein of interest (e.g., a protein with a detectable label such as a fluorescent moiety or a biologically active protein such as a therapeutice protein) into target cells.


In some embodiments, the polypeptide of this invention is linked to a heterologous moiety. This moiety in some instances may be a peptide, such as a reporter protein (which is capable of generating a detectable signal, e.g., a fluorophore peptide such as mCherry), or a therapeutic protein (which is capable of conferring a therapeutic effect when administered), or a transcription factor (which is capable of modulating gene expression, e.g., OCT4 for cell reprogramming), or a gene-editing protein (which is capable of recognizing and cleaving specific DNA sequence, e.g., the RNA-guided CRISPR-associated (Cas) protein and Cre recombinase), or an enzyme (which is capable of recognizing and cleaving specific peptide sequence or structural element or synthetic substrate, such as sortase and cytosine deaminase), thus forming a fusion protein of the modified Cry protein (e.g., modified SEQ ID NO:1 such as SEQ ID NO:2 or a fragment thereof, for example, the 389-471 segment of SEQ ID NO:2) and the peptide, although in other instances the heterologous moiety may be a non-peptide moiety such as a detectable label (e.g., a composition comprising a radioisotope, fluorescent dye, or electron-dense reagent) or a solid substrate/support. In some embodiments, the polypeptide is a fusion protein comprising the modified SEQ ID NO:1 and the therapeutic protein, in which all amino acids at residues 391, 395, 423, 430, 432, 433, 436, 461, 462, 463, and 466 of SEQ ID NO:1 have been replaced with lysine. In some embodiments, the polypeptide as a fusion protein has additional modifications in SEQ ID NO:1, for example, mutations including insertions, deletions, or substitutions at one, optionally two or all three residues 533, 535, and 536. In some embodiments, the fusion protein includes the modified SEQ ID NO:1 comprising amino acids at residues 391, 395, 423, 430, 432, 433, 436, 461, 462, 463, and 466 having been replaced with lysine, and amino acids at residues 533, 535, and 536 having been replaced with alanine, plus a therapeutic protein, such as the p16 protein or the p53 protein. In some embodiments, the fusion protein includes a fragment of a modified Cry protein (e.g., the 389-471 segment of SEQ ID NO:2) and its fusion partner, a heterologous peptide, which may be a protein capable of exerting a detectable signal or a desired biological activity, exemplified above and herein.


In a second aspect, the present invention provides a polynucleotide sequence encoding the polypeptide or fusion protein of this invention as described above and herein. In some embodiments, the polynucleotide sequence is present in an expression cassette, which is typically a recombinantly produced nucleotide structure comprising a promoter (for example, a heterologous promoter) operably linked to the polynucleotide sequence encoding the polypeptide. In some embodiments, the expression cassette may be present in the form of a polynucleotide vector, such as a plasmid or a viral vector. In a related aspect, this invention provides a host cell comprising the polypeptide described above and herein, a host cell comprising the polynucleotide sequence encoding the polypeptide, and a host cell comprising the expression cassette or vector that contains the polynucleotide sequence encoding the polypeptide. In some cases, the host cell is a bacterial cell or one derived from a bacterium, especially a cell of a Bacillus sp. bacterium, such as Bacillus subtilis (Bs) or Bacillus thuringiensis (Bt) cell. In some embodiments, the bacterium is E. coli.


In a third aspect, the present invention provides a method for recombinantly producing the polypeptide or fusion protein of this invention. The method includes the steps of (i) introducing the polynucleotide sequence encoding the polypeptide of this invention as described above and herein into a host cell; and (ii) culturing the cell under conditions permissible for the expression of the polypeptide. The polynucleotide sequence encoding the polypeptide may be in the form of an expression cassette or a vector such as a plasmid. In some embodiments, the host cell expressing the polypeptide of this invention is a bacterial cell, especially of Bacillus sp. such as a Bacillus subtilis (Bs) cell or Bacillus thuringiensis (Bt) cell. Another bacterial strain, such as E. coli, may also be used. In some cases, the method of recombinantly producing the polypeptide further includes a step (iii) of purifying the polypeptide after it has been expressed by the host cell, for example, when the polypeptide is in the crystal form. Typically, the fusion protein assumes a crystalline form or crystalized form upon its expression within the host cells. It may be purified in the crystal form; or it may be purified and then solubilized if necessary.


In a fourth aspect, the present invention provides a composition comprising the polypeptide or fusion protein described above or herein and a mammalian cell. In some embodiments, the polypeptide is crystalized. In some embodiments, the fusion protein is a soluble protein comprising a segment of a modified Cry protein (e.g., the 389-471 segment of SEQ ID NO:2) and its fusion partner, a heterologous protein of desirable properties such as in detectability or a specific biological activity as described above and herein. In some embodiments, the mammalian cell is a cancer cell. In some embodiments, the mammalian cell is an epithelial cell, fibroblast cell, neuronal cell, or immune cell.


In a fifth aspect, the present invention provides a method for delivering an effector protein (such as a therapeutic protein) into a mammalian cell. The method includes the step of contacting the polypeptide or fusion protein of this invention as described above and herein with the mammalian cell, wherein the polypeptide is a fusion protein of a modified Cry protein (e.g., the modified SEQ ID NO:1) and the effector protein, and the polypeptide is crystalized. In the alternative, the fusion protein is a soluble protein comprising a segment of a modified Cry protein (e.g., the 389-471 segment of SEQ ID NO:2) and its fusion partner, a heterologous protein of desirable properties such as in the detectability or a specific biological activity as described above and herein, with the segment of modified Cry protein acting in a manner similar to that of a cell-penetrating peptide to facilitate and enhance the efficiency of transportation of the heterologous protein into target cells.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Cellular uptake of Cry3Aa crystals by A549 cells. Confocal images showing that Cry3Aa crystals cannot be taken up by non-phagocytic A549 cells. grey: Nucleus stained with Hoechst33342 dye; bright white: Alexa 660-labelled Cry3Aa crystals.



FIG. 2. Sequences of the domain II of Cry3Aa and Pos3Aa. Triangles indicate the mutated amino acids.



FIG. 3. Production of Pos3Aa protein crystals and the structure determination of Pos3Aa protein. (A) SDS-PAGE analysis of the purified Pos3Aa protein crystals. (B) SEM images showing the identical size and shape of Pos3Aa crystals to Cry3Aa crystals. (C) Structure of Pos3Aa protein. Mutated amino acids are highlighted with red. (D) Structural alignment of Cry3Aa and Pos3Aa. (E) The surface electrostatic potentials of Cry3Aa (upper panel) and Pos3Aa (lower panel). Dark grey: −10 kT/e; Black: +10 kT/e.



FIG. 4. The flexibility of lysine residues in domain II of Pos3Aa. (A) Low electron density of these lysine residues. (B) High B-factor of mutated lysine residues. Black: 16.53; Light grey: 104.96.



FIG. 5. Efficient cellular uptake of Pos3Aa crystals by non-phagocytic cells and the mechanism. (A) The structure of Pos3Aa crystals. The dotted circles indicate the clustered lysine residues. (B) Confocal images showing the cellular uptake of Pos3Aa crystals by A549, PPF and PC12 cells. (C) Confocal images and (D) flow cytometric analysis showing that no Pos3Aa uptake under 4° C. (E) The effects of different inhibitors on the uptake of Pos3Aa crystals by A549 cells. All three inhibitors for macropinocytosis can block the internalization of Pos3Aa crystals, indicating that the main pathway, by which the Pos3Aa crystals enter non-phagocytic cells is macropinocytosis.



FIG. 6. Endosomal escape of Pos3Aa protein crystals. (A) Representative confocal images showing the localization of Pos3Aa protein crystals (green) in different cells after internalization. (B) Representative confocal images showing the localization of Cry3AaTATHA2 protein crystals (green) in A549 cells after internalization. Fluorescence intensity profiles of the white line area of the merged images in (C) FIG. 5A and (D) FIG. 5B. Nucleus was stained with Hoechst 33342 (blue). Endo/lysosomes were stained with LysoTracker Red DND-99 (red). The bright white color indicates the overlap of protein crystals (green) with endo/lysosomes (red). The bright white color in merged images indicates protein crystals in the cytoplasm.



FIG. 7. Intracellular delivery of mCherry protein by Pos3Aa-mCherry protein crystals. (A) Representative confocal images showing cellular uptake of Pos3Aa-mCherry crystals by A549 cells. (B) Flow cytometric histogram and (C) fold change in mCherry fluorescence intensity of A549 cells showing the cellular uptake of mCherry, TAT-mCherry, R9-mCherry and Pos3Aa-mCherry. (D) Representative confocal images showing the subcellular location of internalized TAT-mCherry, R9-mCherry and Pos3Aa-mCherry in A549 cells, lysosomes were labelled with LysoTracker Green DND-26. (E-G) Fluorescent intensity profiles of the area marked with white boxes in 7D. (E) Pos3Aa-mCherry, (F) R9-mCherry, (G) TAT-mCherry.



FIG. 8. Cellular uptake of Pos3Aa-p53 crystals by MDA-MB-231 cells. Blue: Nucleus stained with Hoechst 33342 dye. Green: Alexa 488-labelled Pos3Aa-p53 crystals. Red: Cell membrane.



FIG. 9. Intracellular delivery of p53 protein by Pos3Aa-p53 protein crystals. (A) Representative confocal images showing cellular uptake of Pos3Aa-mCh-p53 crystals by MDA-MB-231 cells. (B) The mean mCherry fluorescence intensity of MDA-MB-231 cells after the treatment of mCh-p53 protein and Pos3Aa-mCh-p53 crystals showing the dose-dependent uptake of Pos3Aa-mCh-p53. (C) Western blot detection of the Pos3Aa-p53 delivered to the nucleus of MDA-MB-231 cells. (D) Quantitation of the delivered p53 protein in nucleus. (E) Pos3Aa-p53 treatment inhibited the growth of p53-deficient MDA-MB-231 cells. (G) % caspase-3/7 activated cells and (H) relative Caspase-3/7 green fluorescence intensities showing the increased caspase-3/7 activity of cells treated with Pos3Aa-p53 crystals. (I) Cell cycle distributions and (J) % cells in G1 phase showing that Pos3Aa-p53 crystals induced the G1 cell cycle arrest of MDA-MB-231 cells.



FIG. 10. Synergistic inhibition of Pos3Aa-p53 and 5-FU on the growth of MDA-MB-231 cells. MDA-MB-231 cells were pre-treated with 500 nM Pos3Aa-p53 crystals and re-seeded to 96 well plates. Cells with or without pre-treatment were then incubated with a graded dose of (A) 5-FU or (B) Dox for 3 days. (C) MDA-MB-231 cells were treated with 5-FU only or 5-FU and Pos3Aa-p53 crystals for 3 days. Cell viability was determined by MTS reagent.



FIG. 11. Release of mCherry from Pos3Aa-mCherry and Pos3Aa™-mCherry crystals.



FIG. 12. Cellular uptake of Pos3Aa™-mCherry crystals by A549 cells. Blue: nucleus stained with Hoechst 33342 dye; Red: Pos3Aa™-mCherry crystals; Green: Cell membrane stained with the Wheat Germ Agglutinin Alexa 647 dye.



FIG. 13. Intracellular delivery of p16 protein by Pos3Aa™-mCh-p16 crystals. (A) Cellular uptake of Pos3Aa™-mCh-p16 crystals by UM-SCC-22A cells after 24 h, 48 h, and 96 h incubation. Diffused mCherry signal in both cytoplasm and nucleus indicate the released mCherry proteins. (B) Merged flow cytometric histogram and (C) mean mCherry fluorescence intensity showing the dose-dependent uptake of Pos3Aa™-mCh-p16 crystals by UM-SCC-22A cells. p16 dose-dependent cell cycle arrest of UM-SCC-22A cells. (D) Representative flow cytometric histograms of cell cycle alterations and (E) % cells in G1 phase of UM-SCC-22A cells treated with 500 nM mCh-p16 protein, Pos3Aa™-mCherry and Pos3Aa™-mCh-p16 crystals for 48 h. (F) Delivery of p16 protein reduced the level of phosphorylated Rb in UM-SCC-22A cells.



FIG. 14. The amino acid sequence of P3AP peptide.



FIG. 15. SDS-PAGE analysis of purified P3AP-mCherry protein (indicated by asterisk).



FIG. 16. Cellular uptake of mCherry, R9-mCherry and P3AP-mCherry proteins by MDA-MB-231 cells. Blue: nucleus stained with Hoechst 33342. Red: different proteins. Green: cell membrane stained with Wheat Germ Agglutinin, Alexa Fluor™ 647 Conjugate.



FIG. 17. Uptake efficiency and cytotoxicity of P3AP-mCherry. (A) Representative flow cytometric histograms showing the uptake of mCherry, R9-mCherry, TAT-mCherry and P3AP-mCherry proteins. (B) Quantitated mean mCherry fluorescence intensities. (C) Cell viabilities of MDA-MB-231 cells with different treatments. The inner bar graph shows the cell viabilities at 5 μM protein concentration.



FIG. 18. Structure-based sequence alignments of other three-domain Cry proteins to Cry3Aa protein. Domain II of Cry3Aa protein is highlighted in light grey. Residues in other Cry proteins corresponding to the domain II of Cry3Aa protein are highlighted in dark grey.





DEFINITIONS

The term “Cry protein,” as used herein, refers to any one protein among a class of crystalline three-domain Cry proteins produced by strains of Bacillus thuringiensis (http://www.lifesci.sussex.ac.uk/home/Neil_Crickmore/Bt/). Some examples of “Cry proteins” include, but are not limited to, Cry1Aa, Cry2Aa, Cry3Aa, Cry4Aa, Cry5B, Cry7Ca1, Cry8Ea1, Cry10Aa, and Cry11Aa. Their amino acid sequences and polynucleotide coding sequences are known (set forth in SEQ ID NOs:1 and 4-11). Their GenBank Accession Numbers are:


















Cry1Aa
AAA22353.1



Cry2Aa
AAA22335.1



Cry3Aa
AAA22541.1



Cry4Aa
CAA68485.1



Cry5B
AAA68598.1



Cry7Ca1
ABR67863.1



Cry8Ea1
AY329081.1



Cry10Aa
AAA22614.1



Cry11Aa
AAA22352.1










In addition to the wild-type Cry proteins, the term “Cry protein” also encompasses functional variants, which (1) share an amino acid sequence identity of at least 80%, 81%, 82%, 83%, 84%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5% to the polypeptide sequence of any one of the three-domain Cry proteins listed in http://www.lifesci.sussex.ac.uk/home/Neil_Crickmore/Bt/; and (2) retain the ability to spontaneously form crystals within host cells as can be confirmed by known methods such as electron micrograph (see description in, e.g., Park et al., Appl Environ Microbiol, 1998, 64, 3932-3938; Schnepf et al., Microbiol Mol Biol Rev, 1998, 62, 775-806; Whiteley and Schnepf, Annu Rev Microbiol, 1986, 40, 549-576; and Nair et al., PLoS One, 2015, 10, e0127669).


The term “nucleic acid” or “polynucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)). The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.


The term “gene” means the segment of DNA involved in producing a polypeptide chain. It may include regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).


The term “amino acid” refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, γ-carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an α carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. “Amino acid mimetics” refers to chemical compounds having a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.


There are various known methods in the art that permit the incorporation of an unnatural amino acid derivative or analog into a polypeptide chain in a site-specific manner, see, e.g., WO 02/086075.


Amino acids may be referred to herein by either the commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.


“Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids that encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid that encodes a polypeptide is implicit in each described sequence.


As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.


The following eight groups each contain amino acids that are conservative substitutions for one another:

  • 1) Alanine (A), Glycine (G);
  • 2) Aspartic acid (D), Glutamic acid (E);
  • 3) Asparagine (N), Glutamine (Q);
  • 4) Arginine (R), Lysine (K);
  • 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);
  • 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);
  • 7) Serine (S), Threonine (T); and
  • 8) Cysteine (C), Methionine (M)


    (see, e.g., Creighton, Proteins, W. H. Freeman and Co., N. Y. (1984)).


Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.


In the present application, amino acid residues are numbered according to their relative positions from the left most residue, which is numbered 1, in an unmodified wild-type polypeptide sequence.


As used in herein, the terms “identical” or percent “identity,” in the context of describing two or more polynucleotide or amino acid sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (for example, a Cry protein or a modified Cry protein sequence of this invention has at least 80% identity, preferably 85%, 90%, 91%, 92%, 93, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity, to a reference sequence, e.g., the amino acid sequence of a corresponding wild-type Cry protein), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be “substantially identical.” With regard to polynucleotide sequences, this definition also refers to the complement of a test sequence. Preferably, the identity exists over a region that is at least about 50 amino acids or nucleotides in length, or more preferably over a region that is 75-100 amino acids or nucleotides in length.


For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. For sequence comparison of nucleic acids and proteins, the BLAST and BLAST 2.0 algorithms and the default parameters discussed below are used.


A “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat'l. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Current Protocols in Molecular Biology (Ausubel et al., eds. 1995 supplement)).


Examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al., (1990) J. Mol. Biol. 215: 403-410 and Altschul et al. (1977) Nucleic Acids Res. 25: 3389-3402, respectively. Software for performing BLAST analyses is publicly available at the National Center for Biotechnology Information website, ncbi.nlm.nih.gov. The algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits acts as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a word size (W) of 28, an expectation (E) of 10, M=1, N=−2, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a word size (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989)).


The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul, Proc. Nat'l. Acad. Sci. USA 90:5873-5787 (1993)). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.


An indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below. Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.


“Polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. All three terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bonds.


The term “recombinant” when used with reference, e.g., to a cell, or a nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.


A “promoter” is defined as an array of nucleic acid control sequences that direct transcription of a polynucleotide sequence. As used herein, a promoter includes necessary polynucleotide sequences near the start site of transcription, such as, in the case of a polymerase II type promoter, a TATA element. A promoter also optionally includes distal enhancer or repressor elements, which can be located as much as several thousand base pairs from the start site of transcription. A “constitutive” promoter is a promoter that is active under most environmental and developmental conditions. An “inducible” promoter is a promoter that is active under environmental or developmental regulation. The term “operably linked” refers to a functional linkage between a polynucleotide expression control sequence (such as a promoter, or array of transcription factor binding sites) and a second polynucleotide sequence, wherein the expression control sequence directs transcription of the polynucleotide sequence corresponding to the second sequence.


An “expression cassette” is a nucleic acid construct, generated recombinantly or synthetically, with a series of specified polynucleotide elements that permit transcription of a particular polynucleotide sequence in a host cell. An expression cassette may be part of a plasmid, viral genome, or nucleic acid fragment. Typically, an expression cassette includes a polynucleotide to be transcribed, operably linked to a promoter.


The term “heterologous” as used in the context of describing the relative location of two elements, refers to the two elements such as polynucleotide sequences (e.g., a promoter or a protein/polypeptide-encoding sequence) or polypeptide sequences (e.g., a modified Cry protein of this invention or a fusion protein comprising such a modified Cry protein) that are not naturally found in the same relative positions. Thus, a “heterologous promoter” of a gene refers to a promoter that is not naturally operably linked to that gene. Similarly, a “heterologous polypeptide” or “heterologous polynucleotide” to a modified Cry protein or its encoding sequence is one derived from an origin other than this particular Cry protein in the wild-type version, or one derived from the wild-type Cry protein but the fusion of a modified Cry protein (or its coding sequence) with a heterologous polypeptide (or polynucleotide sequence) does not result in a longer polypeptide or polynucleotide sequence that can be found naturally in the corresponding wild-type Cry protein (or its coding sequence).


A “label,” “detectable label,” or “detectable moiety” is a composition detectable by radiological, spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means. For example, useful labels include radioisotopes such as 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins that can be made detectable, e.g., by incorporating a radioactive component into a polypeptide or used to detect antibodies specifically reactive with the polypeptide. Typically a detectable label is a heterologous moiety attached to a probe or a molecule (e.g., a protein or nucleic acid) with defined binding characteristics (e.g., a polypeptide with a known binding specificity or a polynucleotide), so as to allow the presence of the probe/molecule (and therefore its binding target) to be readily detectable. The heterologous nature of the label ensures that it has an origin different from that of the probe or molecule that it labels, such that the probe/molecule attached with the detectable label does not constitute a naturally occurring composition.


A “host cell” is a cell that contains an expression vector and supports the replication or expression of the expression vector. Host cells may be prokaryotic cells such as E. coli or Bacillus thuringiensis, or eukaryotic cells such as yeast, insect, amphibian, or mammalian cells such as CHO, HeLa and the like, e.g., cultured cells, explants, and cells in vivo.


The term “about” as used herein denotes a range of +/−10% of a reference value. For examples, “about 10” defines a range of 9 to 11.


DETAILED DESCRIPTION OF THE INVENTION
I. Introduction

There has been growing interest in devising new and more effective methods for administration of therapeutic proteins for the purpose of treating medical conditions and disorders. By generating modified Cry proteins capable of self-crystallization and their fragments capable of enhancing protein intake across cell membrane, the present inventors have developed an innovative and effective strategy to deliver therapeutic proteins.


II. Production of Modified Cry Proteins and Their Fragments

A. General Recombinant Technology


Basic texts disclosing general methods and techniques in the field of recombinant genetics include Sambrook and Russell, Molecular Cloning, A Laboratory Manual (3rd ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Ausubel et al., eds., Current Protocols in Molecular Biology (1994).


For nucleic acids, sizes are given in either kilobases (kb) or base pairs (bp). These are estimates derived from agarose or acrylamide gel electrophoresis, from sequenced nucleic acids, or from published DNA sequences. For proteins, sizes are given in kilodaltons (kDa) or amino acid residue numbers. Proteins sizes are estimated from gel electrophoresis, from sequenced proteins, from derived amino acid sequences, or from published protein sequences.


Oligonucleotides that are not commercially available can be chemically synthesized, e.g., according to the solid phase phosphoramidite triester method first described by Beaucage & Caruthers, Tetrahedron Lett. 22: 1859-1862 (1981), using an automated synthesizer, as described in Van Devanter et. al., Nucleic Acids Res. 12: 6159-6168 (1984). Purification of oligonucleotides is performed using any art-recognized strategy, e.g., native acrylamide gel electrophoresis or anion-exchange HPLC as described in Pearson & Reanier, J. Chrom. 255: 137-149 (1983).


The sequence of a gene of interest, such as the polynucleotide sequence encoding a modified Cry protein or fusion protein thereof, and synthetic oligonucleotides can be verified after cloning or subcloning using, e.g., the chain termination method for sequencing double-stranded templates of Wallace et al., Gene 16: 21-26 (1981).


B. Coding Sequence for a Modified Cry Protein


Polynucleotide sequences encoding modified Cry proteins, their fragments, or fusion proteins of this invention can be readily constructed by modifying a wild-type Cry protein to obtain a variant or fragment and optionally combining the coding sequences for the fusion partners, such as a Cry3Aa protein and p53 or p16 protein. The sequences for Cry proteins and enzymes are generally known and may be obtained from a commercial supplier.


In addition to the use of full length wild-type Cry proteins for constructing the modified Cry proteins or fusion proteins of this invention, fragments of Cry proteins and/or variants of Cry proteins may also be useful. A DNA sequence encoding a Cry protein can be modified to generate fragments or variants of the Cry protein. So long as the fragments and variants retain the ability to spontaneously form crystals when expressed in a host cell, especially a Bacillus bacterial cell, they can be used for producing the fusion proteins and render the fusion proteins the ability to undergo spontaneous crystallization. In some cases, soluble fragments corresponding to domain II of a Cry protein (including a modified version) such as P3AP are developed for use in the enhanced delivery of proteins having desired activity or function to target cells. Typically, the variants bear a high percentage of sequence identity (e.g., at least 80, 85, 90, 95, 97, 98, 99% or higher) to the wild-type Cry protein sequence, whereas the fragments may be substantially shorter than the full length Cry protein, such as having some amino acids (e.g., 10-300 or 20-200 or 50-100 amino acids) removed from the N- or C-terminus of the full length Cry protein. For example, a useful Cry3Aa fragment may be as short as the first 290 amino acids from the N-terminus, encompassing Domain I of the protein. Other examples of such fragments include a Cry protein fragment having its first 57 amino acids from N-terminus removed and a Cry protein fragment having its C-terminal 18 amino acids removed. The ability of a modified Cry protein or a fusion protein thereof to undergo spontaneous crystallization can be verified by electron micrograph, whereas the desired biological activity attributable to the fusion partner or the “cargo” protein (e.g., cancer-suppressing protein such as p53 or p16) can be confirmed by established assays for each specific “cargo” protein. Surprisingly, the present inventors discovered during their studies that the presence of a modified Cry protein having multiple lysines introduced into domain II in a fusion protein affords a significant increase in the cellular uptake and endosomal escape of the fusion protein, thus permitting more effective delivery of therapeutic proteins to target cells (e.g., cancer cells). The inventors further revealed that modification to the Cry protein in domain III is able to confer to the fusion protein significantly enhanced solubility, providing additional enhancement to therapeutic efficacy of the “cargo” protein.


In addition, the present inventors discovered that a fragment of a modified Cry3Aa protein, such as a fragment of Pos3Aa (SEQ ID NO:2) generally corresponding to domain II of the protein, i.e., starting at residue 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, or 394 and ending at residue 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, or 486 of SEQ ID NO:2, although a soluble peptide, is capable of efficiently crossing cell membrane and therefore can be used to effectively deliver a protein of interest (e.g., a protein with desired biological function or activity) into target cells when peptide is fused to the protein. P3AP, the 389-471 segment of SEQ ID NO:2, is an exemplary peptide of the present invention useful for such applications.


In some cases, a peptide linker or spacer is used between the coding sequences for the modified Cry protein or its fragment and its fusion partner, a heterologous protein or an effector protein. Such heterologous protein may be of any nature and any size, although in some cases it is one within the molecular weight range of about 2-200 kDa, or about 5-100 or 10-100 kDa, or about 15-75 kDa. In particular, proteins that are hard to produce (e.g., p16 protein) or easy to aggregate when produced in E. coli or any other protein production system (e.g., p53 protein) may still suitable for use in this delivery method. One purpose is to ensure the proper reading frame for the fusion protein such that the coding sequences for both modified Cry protein or fragment and the heterologous protein are in frame. Another purpose is to provide appropriate spatial relationship between the modified Cry protein and the heterologous protein, such that each may retain its original functionality: the modified Cry protein is able to cause self-crystallization of the fusion protein, and the heterologous protein remains active in its desirable biological activity (e.g., cancer-suppressing capacity). Also, one or more linkers may be placed at the very beginning and/or the very end of the open reading frame, so as to facilitate proper start and termination of the coding sequence translation. Such linkage amino acid sequences are usually shorts and typically no longer than 100 or 50 amino acids, such as between 1 to 100, 1 or 2 to 50, 2 or 3 to 25, 3 or 4 to 10 amino acids.


C. Sequence Modification for Preferred Codon Usage in a Host Organism


The polynucleotide sequence encoding a modified Cry protein or a fragment thereof or fusion protein of this invention can be further altered to coincide with the preferred codon usage of a particular host. For example, the preferred codon usage of one strain of bacterial cells can be used to derive a polynucleotide that encodes a recombinant polypeptide of the invention and includes the codons favored by this strain. The frequency of preferred codon usage exhibited by a host cell can be calculated by averaging frequency of preferred codon usage in a large number of genes expressed by the host cell (e.g., calculation service is available from web site of the Kazusa DNA Research Institute, Japan). This analysis is preferably limited to genes that are highly expressed by the host cell.


At the completion of modification, the coding sequences are verified by sequencing and are then subcloned into an appropriate expression vector for recombinant production of a modified Cry protein or its fragment or a fusion protein thereof.


III. Expression and Purification of Modified Cry Proteins or Fusion Proteins

Following verification of the coding sequence, a modified Cry protein/its fragment or fusion protein of this invention can be produced using routine techniques in the field of recombinant genetics, relying on the polynucleotide sequences encoding the modified Cry protein/its fragment or fusion protein disclosed herein.


A. Expression Systems


To obtain high level expression of a nucleic acid encoding a recombinant protein of this invention, one typically subclones a polynucleotide encoding the protein in the correct reading frame into an expression vector that contains a strong promoter to direct transcription, a transcription/translation terminator and a ribosome binding site for translational initiation. Suitable bacterial promoters are well known in the art and described, e.g., in Sambrook and Russell, supra, and Ausubel et al., supra. Bacterial expression systems for expressing the polypeptide are available in, e.g., E. coli, Bacillus sp., Salmonella, and Caulobacter. Kits for such expression systems are commercially available.


The promoter used to direct expression of a heterologous nucleic acid depends on the particular application. The promoter is optionally positioned about the same distance from the heterologous transcription start site as it is from the transcription start site in its natural setting. As is known in the art, however, some variation in this distance can be accommodated without loss of promoter function. In some cases, a constitutive promoter is used, whereas in other cases an inducible promoter rather than a constitutive promoter is preferred.


In addition to the promoter, the expression vector typically includes a transcription unit or expression cassette that contains all the additional elements required for the expression of the polypeptide of this invention in host cells. A typical expression cassette thus contains a promoter operably linked to the nucleic acid sequence encoding the recombinant protein and signals required for efficient polyadenylation of the transcript, ribosome binding sites, and translation termination. The nucleic acid sequence encoding the recombinant protein may be linked to a cleavable signal peptide sequence to promote secretion of the polypeptide by the transformed cell. Such signal peptides include, among others, the signal peptides from tissue plasminogen activator, insulin, and neuron growth factor, and juvenile hormone esterase of Heliothis virescens. Additional elements of the cassette may include enhancers and, if genomic DNA is used as the structural gene, introns with functional splice donor and acceptor sites.


In addition to a promoter sequence, the expression cassette should also contain a transcription termination region downstream of the coding sequence to provide for efficient termination. The termination region may be obtained from the same gene as the promoter sequence or may be obtained from different genes.


The particular expression vector used to transport the genetic information into the cell is not particularly critical. Any of the conventional vectors used for expression in eukaryotic or prokaryotic cells may be used, especially those suitable for expression in cells of Bacillus sp. such as Bt and Bs. Standard bacterial expression vectors include plasmids such as pBR322 based plasmids, pSKF, pET23D, and fusion expression systems such as GST and LacZ.


The elements that are typically included in expression vectors also include a replicon that functions in bacteria such as Bacillus sp. and E. coli, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of coding sequences. The particular antibiotic resistance gene chosen is not critical, any of the many resistance genes known in the art are suitable. Similar to antibiotic resistance selection markers, metabolic selection markers based on known metabolic pathways may also be used as a means for selecting transformed host cells.


B. Transfection Methods


Standard transfection methods are used to produce bacterial, mammalian, yeast, insect, or plant cell lines that express large quantities of a recombinant protein of this invention, which are then purified using standard techniques (see, e.g., Colley et al., J. Biol. Chem. 264: 17619-17622 (1989); Guide to Protein Purification, in Methods in Enzymology, vol. 182 (Deutscher, ed., 1990)). Transformation of eukaryotic and prokaryotic cells are performed according to standard techniques (see, e.g., Morrison, J. Bact. 132: 349-351 (1977); Clark-Curtiss & Curtiss, Methods in Enzymology 101: 347-362 (Wu et al., eds, 1983).


Any of the well-known procedures for introducing foreign nucleotide sequences into host cells may be used. These include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, liposomes, microinjection, plasma vectors, viral vectors and any of the other well-known methods for introducing cloned genomic DNA, cDNA, synthetic DNA, or other foreign genetic material into a host cell (see, e.g., Sambrook and Russell, supra). It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing at least one gene into the host cell capable of expressing the recombinant protein of this invention.


C. Purification of Modified Cry Proteins and Fusion Proteins


Once the expression of a modified Cry protein or its fragment or a fusion protein thereof in transfected host cells is confirmed, e.g., via electron micrograph for detecting protein crystals or an immunoassay such as Western blotting analysis, the host cells are then cultured in an appropriate scale for the purpose of purifying the recombinant protein.


When the Cry fusion proteins or fusion proteins of the present invention are produced recombinantly by transformed bacteria in large amounts, for example after promoter induction, the proteins are present in crystalline form or insoluble aggregates within the host cells. Thus, one can readily isolate the crystals from the cell lysate based on their distinct density by utilizing techniques such as centrifugation and density gradient separation followed by one or more rinsing steps to further remove contaminants from the protein crystals.


There are several protocols that are suitable for purification of protein inclusion bodies. For example, purification of aggregate proteins (hereinafter referred to as inclusion bodies) typically involves the extraction, separation and/or purification of inclusion bodies by disruption of bacterial cells, e.g., by incubation in a buffer of about 100-150 μg/ml lysozyme and 0.1% Nonidet P40, a non-ionic detergent. The cell suspension can be ground using a Polytron grinder (Brinkman Instruments, Westbury, N.Y.). Alternatively, the cells can be sonicated on ice. Additional methods of lysing bacteria are described in Ausubel et al. and Sambrook and Russell, both supra, and will be apparent to those of skill in the art.


The cell suspension is generally centrifuged and the pellet containing the inclusion bodies resuspended in buffer which does not dissolve but washes the inclusion bodies, e.g., 20 mM Tris-HCl (pH 7.2), 1 mM EDTA, 150 mM NaCl and 2% Triton-X 100, a non-ionic detergent. It may be necessary to repeat the wash step to remove as much cellular debris as possible. The remaining pellet of inclusion bodies may be resuspended in an appropriate buffer (e.g., 20 mM sodium phosphate, pH 6.8, 150 mM NaCl). Other appropriate buffers will be apparent to those of skill in the art.


Following the washing step, the inclusion bodies are solubilized by the addition of a solvent that is both a strong hydrogen acceptor and a strong hydrogen donor (or a combination of solvents each having one of these properties). The proteins that formed the inclusion bodies may then be renatured by dilution or dialysis with a compatible buffer. Suitable solvents include, but are not limited to, urea (from about 4 M to about 8 M), formamide (at least about 80%, volume/volume basis), and guanidine hydrochloride (from about 4 M to about 8 M). Some solvents that are capable of solubilizing aggregate-forming proteins, such as SDS (sodium dodecyl sulfate) and 70% formic acid, may be inappropriate for use in this procedure due to the possibility of irreversible denaturation of the proteins, accompanied by a lack of immunogenicity and/or activity. Although guanidine hydrochloride and similar agents are denaturants, this denaturation is not irreversible and renaturation may occur upon removal (by dialysis, for example) or dilution of the denaturant, allowing re-formation of the immunologically and/or biologically active protein of interest. After solubilization, the protein can be separated from other bacterial proteins by standard separation techniques. For further description of purifying recombinant polypeptides from bacterial inclusion body, see, e.g., Patra et al., Protein Expression and Purification 18: 182-190 (2000).


While the Cry fusion protein crystals tend to remain insoluble at lower or neutral pHs, placing them in alkaline solutions with pH at or greater than 10 or 11 can often effectively dissolve the protein. Once dissolved, the protein can then be analyzed by gel separation (e.g., on an SDS gel) and immunoassays to confirm its identity based on the appropriate molecular weight and immunoreactivity.


D. Crosslinking Cry Fusion Proteins


Crosslinking is a commonly used technique for a broad ranges of goals, such as to stabilize protein tertiary and quaternary structure for analysis; to capture and identify unknown protein interactors or interaction domains; to conjugate an enzyme or tag to an antibody or other purified protein; to immobilize antibodies or other proteins for assays or affinity-purification; and to attach peptides to larger “carrier” proteins to facilitate handling/storage. The present inventors have observed that crosslinking tends to further enhance the desirable properties of the Cry fusion protein crystals such as thermostability and tolerance to organic solvents. Thus, in some cases there is a preference to further crosslink a Cry fusion protein upon its recombinant production and purification.


Despite the complexity of protein structure, including composition with 20 different amino acids, only a small number of protein functional groups comprise selectable targets for practical crosslinking methods. In fact, just four protein chemical targets account for the vast majority of crosslinking and chemical modification techniques: (1) primary amines (—NH2): this group exists at the N-terminus of each polypeptide chain and in the side chain of lysine (Lys, K) residues; (2) carboxyls (—COOH): this group exists at the C-terminus of each polypeptide chain and in the side chains of aspartic acid (Asp, D) and glutamic acid (Glu, E); (3) sulfhydryls (—SH): this group exists in the side chain of cysteine (Cys, C). Often, as part of a protein's secondary or tertiary structure, cysteines are joined together between their side chains via disulfide bonds (—S—S—); and (4) carbonyls (—CHO): these aldehyde groups can be created by oxidizing carbohydrate groups in glycoproteins. For each of these protein functional-group targets, there exist one to several types of reactive groups that are capable of targeting them and have been used as the basis for synthesizing crosslinking and modification reagents. Crosslinkers are selected on the basis of their chemical reactivities (i.e., specificity for particular function groups) and other chemical properties that facilitate their use in different specific applications.


After a fusion protein of the present invention, e.g., a Pos3Aa-p53 fusion protein, is recombinantly produced in host cells (such as Bacillus subtilis cells or Bacillus thuringiensis cells) in a crystalline form and then properly purified, it can then be chemically crosslinked to further increase the level of enhancement in the protein's properties such as thermosstability and tolerance to organic solvents. Well-known chemical crosslinking reagents can be used for this purpose in accordance with the established procedures. Some examples of suitable crosslinking reagents include glutaraldehyde, bis(sulfosuccinimidyl)suberate (BS3), phenol-formaldehyde, Lys to lys cross-linking: DSG (disuccinimidyl glutarate), Lys to cys cross-linking: Sulfo-EMCS (N-ε-maleimidocaproyl-oxysulfosuccinimide ester), Cys to cys cross-linking, and BMH (bismaleimidohexane).


IV. Pharmaceutical Compositions and Administration

The present invention also provides pharmaceutical compositions comprising an effective amount of a modified Cry fusion protein for achieving the intended effect by the biological activity attributable to the fusion partner or “cargo” protein of the modified Cry protein or a fragment thereof (e.g., P3AP), therefore useful in both prophylactic and therapeutic applications depending on the specific target disease and therapeutic protein. Pharmaceutical compositions of the invention are suitable for use in a variety of drug delivery systems. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed. (1985). For a brief review of methods for drug delivery, see, Langer, Science 249: 1527-1533 (1990).


The pharmaceutical compositions of the present invention can be administered by various routes, e.g., oral, subcutaneous, transdermal, transnasal, intramuscular, intravenous, or intraperitoneal. The routes of administering the pharmaceutical compositions include systemic or local delivery to a subject suffering from a neurodegenerative disease at daily doses of about 0.01-5000 mg, preferably 5-500 mg, of a Cry fusion protein for a 70 kg adult human per day. The appropriate dose may be administered in a single daily dose or as divided doses presented at appropriate intervals, for example as two, three, four, or more subdoses per day.


For preparing pharmaceutical compositions containing a Cry fusion protein, inert and pharmaceutically acceptable carriers are used. The pharmaceutical carrier can be either solid or liquid. Solid form preparations include, for example, powders, tablets, dispersible granules, capsules, cachets, and suppositories. A solid carrier can be one or more substances that can also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material.


In powders, the carrier is generally a finely divided solid that is in a mixture with the finely divided active component, e.g., a Cry fusion protein comprising a therapeutic protein such as p53 or p16. In tablets, the active ingredient (the Cry fusion protein) is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.


For preparing pharmaceutical compositions in the form of suppositories, a low-melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient-sized molds and allowed to cool and solidify.


Powders and tablets preferably contain between about 5% to about 70% by weight of the active ingredient. Suitable carriers include, for example, magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low-melting wax, cocoa butter, and the like.


The pharmaceutical compositions can include the formulation of the active ingredient of a Cry fusion protein with encapsulating material as a carrier providing a capsule in which the recombinant polypeptide (with or without other carriers) is surrounded by the carrier, such that the carrier is thus in association with the polypeptide. In a similar manner, cachets can also be included. Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration.


Liquid pharmaceutical compositions include, for example, solutions suitable for oral or parenteral administration, suspensions, and emulsions suitable for oral administration. Sterile water solutions of the active component (e.g., a Cry fusion protein) or sterile solutions of the active component in solvents comprising water, buffered water, saline, PBS, ethanol, or propylene glycol are examples of liquid compositions suitable for parenteral administration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents, and the like.


Sterile solutions can be prepared by dissolving the active component (e.g., a Cry fusion protein) in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 11, more preferably from 5 to 9, and most preferably from 7 to 8.


The pharmaceutical compositions containing the Cry fusion protein can be administered for prophylactic and/or therapeutic treatments. In therapeutic applications, compositions are administered to a patient already suffering from a target condition/disease in an amount sufficient to prevent, cure, reverse, or at least partially slow or arrest the symptoms of the condition and its complications. An amount adequate to accomplish this is defined as a “therapeutically effective dose.” Amounts effective for this use will depend on the severity of the disease or condition and the weight and general state of the patient, but generally range from about 0.1 mg to about 2,000 mg of the recombinant polypeptide per day for a 70 kg patient, with dosages of from about 5 mg to about 500 mg of the recombinant polypeptide per day for a 70 kg patient being more commonly used.


In prophylactic applications, pharmaceutical compositions containing a Cry fusion protein are administered to a patient susceptible to or otherwise at risk of developing a target disease or disorder in an amount sufficient to delay or prevent the onset of the symptoms. Such an amount is defined to be a “prophylactically effective dose.” In this use, the precise amounts of the recombinant polypeptide again depend on the patient's state of health and weight, but generally range from about 0.1 mg to about 2,000 mg of the recombinant polypeptide for a 70 kg patient per day, more commonly from about 5 mg to about 500 mg for a 70 kg patient per day.


Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician. In any event, the pharmaceutical formulations should provide a quantity of a Cry fusion protein sufficient to effectively achieve the intended therapeutic effects, e.g., inhibit cancer cell proliferation, invasion and/or metastasis in the patient, either therapeutically or prophylactically.


V. Kits

The invention also provides kits for prophylactic or therapeutic applications by administering a Cry fusion protein according to the method of the present invention. The kits typically include a first container that contains a pharmaceutical composition having an effective amount of a Cry fusion protein, for example, having a therapeutic protein with anti-cancer activity as the fusion partner with a modified Cry protein or a fragment thereof, optionally with a second container containing a second therapeutically active agent, for example, another anti-cancer agent. In some cases, the kits will also include informational material containing instructions on how to dispense the pharmaceutical composition, including description of the type of patients who may be treated (e.g., a person suffering from a condition or disease suitable for treatment by the fusion partner or “cargo” protein in the Cry fusion protein of this invention), the schedule (e.g., administration dose and frequency), route of administration, and the like.


EXAMPLES

The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results.


BACKGROUND

Proteins perform essential biological functions in cells, such as gene regulation, signal transduction and enzyme catalysis, making them potential candidates/targets for drug research and development. Highlighting the tremendous potential of the protein therapeutics market, seven of the top ten drugs sold globally in 2018 were monoclonal antibodies, with numerous other proteins being explored for the treatment of various diseases. While almost all current approved protein-based drugs in the market act on extracellular targets, many diseases are caused by the dysfunction of intracellular proteins. The lack of protein-based therapeutics in the clinic is due in large part to the instability and low cell penetration efficiency of these therapeutics into cells. Moreover, entrapment by endosomes and lysosomes after cell entry can dramatically hinder the efficacy of these proteins. To address these issues, multiple strategies, including cell membrane deformation, hypertonic buffer treatment, cell penetrating peptides and nano/micro-carriers, have been explored to mediate the internalization of protein therapeutics into mammalian cells. Considering the protection to cargo proteins and the versatility for in vivo application, nano/micro-carriers are arguably one of the most promising approaches for protein delivery. Various materials, like cationic polymers, lipids, inorganic materials and proteins, have been utilized to manufacture nano/micro-carriers with different chemical and physical properties. Compared to other materials, protein-based particles have distinct features well suited for intracellular drug delivery, including good biocompatibility, biodegradability, ease of modification.


It was previously reported the development of a novel protein delivery platform based on Cry3Aa protein that naturally forms sub-micrometer-sized protein crystals within the bacterium Bacillus thuringiensis (Bt). In mouse studies, the purified Cry3Aa crystals exhibited great biocompatibility and biodegradability, and minimal toxicity. More importantly, this platform appears to stabilize its cargo proteins in the form of Cry3Aa-cargo fusion protein crystals as demonstrated by the extended lifetime of the protein compared to free protein. It was further shown that these protein crystals could be specifically taken up by phagocytic macrophages. As proof of concept, an antimicrobial peptide has been encapsulated in high loading efficiency into Cry3Aa crystals and successfully delivered to macrophages for treating intracellular parasites infection in a mouse model of cutaneous leishmaniasis. To expand the application of this platform on other non-phagocytic cells, a positively-charged mutant of Cry3Aa (Pos3Aa), which retains its crystal-forming ability in Bt cells, was identified. In this disclosure, the Pos3Aa protein crystals are demonstrated to be readily taken up by various types of non-phagocytic cells, such as cancer cells and fibroblasts, with high efficiency of endosomal escape. Successful intracellular delivery of a fluorescent mCherry protein and two tumor suppressor proteins p53 and p16 was achieved via this Pos3Aa platform. Significantly, Pos3Aa-mediated p53 and p16 delivery restored their anti-cancer activities in p53 or p16-deficient cancer cells, indicating that Pos3Aa-based protein crystals can be an effective platform for intracellular protein delivery.


Results
1. Design and Structure of Pos3Aa Protein

Positively charged amino acids (lysine and arginine) are proven to be crucial for the efficient cellular uptake of cell-penetrating peptides or supercharged proteins by mammalian cells. As above-mentioned, it was discovered that wildtype Cry3Aa protein harbor a large proportion of negatively charged surface residues in the domain II, which is presumably correlated to the poor internalization of Cry3Aa protein crystals into non-phagocytic cells (FIG. 1). It was accordingly hypothesized that the substitutions of these surface-exposed glutamic acid (E), aspartic acid (D) or other polar amino acids to lysine might improve the uptake efficiency of Cry3Aa crystals by non-phagocytic cells. Hence, eleven solvent-exposed amino acids with flexible residues in domain II were identified based on the structure of Cry3Aa protein and mutated to lysine (FIG. 2). The resultant Cry3Aa mutant protein, named Pos3Aa, still formed protein crystals with identical size and morphology to wildtype Cry3Aa when expressed in Bt cells (FIG. 3A, B), suggesting that these mutations may not affect the protein folding and crystal packing. To confirm this, the in vitro formed crystals of Pos3Aa protein were obtained using sitting drop vapor diffusion crystallization and its structure was determined using X-ray crystallography. Pos3Aa crystal shared the same space group (C2221) as Cry3Aa, and Pos3Aa protein maintained the typical three domains structure of Cry toxins (FIG. 3C). Alignment of Pos3Aa to Cry3Aa showed a Ca RMSD value of 0.423 Å, revealing a high structural similarity (FIG. 3D). While the lysine mutations did not abolish the native structure of Cry3Aa protein, the dramatic change in surface electrostatic potential from negative to positive in domain II region was observed consistent with our expectation (FIG. 3E). Moreover, it was discovered that these lysine residues formed three protruding lysine clusters with excessive flexibility, which is reflected in their low electron density and high B-factor (FIG. 4). Previous studies have shown that flexible and clustered lysine residues are critical for the phospholipid binding and membrane permeability, supporting the notion that Pos3Aa protein crystals might be able to interact with the cell membrane and be internalized by non-phagocytic cells.


2. Cellular Uptake of Pos3Aa Crystals and its Mechanism

Pos3Aa protein crystals were produced in Spo407-OA Bt cells, purified by sucrose gradient centrifugation and characterized for the subsequent experiments. Interestingly, Pos3Aa crystals hold a zeta potential value of −14.9 mV, indicating a near-neutral surface of these crystals instead of a positively charged one. One possible explanation, based on the structure of Pos3Aa crystals, is that the lysine clusters are regularly distributed on the limited regions of the crystal surface due to the crystal packing, and meanwhile other regions are still neutral or negatively charged (FIG. 5A).


To evaluate the cellular uptake efficiency of Pos3Aa protein crystals, Alexa488-labeled Pos3Aa crystals (Alexa488-Pos3Aa) were incubated with A549 cells, primary pulmonary fibroblasts (PPFs) and PC12 cells for 24 h and the internalization of crystals was examined using fluorescent confocal microscopy. As indicated in FIG. 5B, Pos3Aa protein crystals were readily taken up by different type of non-phagocytic cells, which wild type Cry3Aa crystals cannot enter, confirming the importance of the clustered lysine residues on determining the cellular uptake potency. To further understand the mechanisms by which Pos3Aa crystals enter these cells, 500 nM Alexa488-Pos3Aa crystals were incubated with A549 cells at 37° C. and 4° C., respectively, for 6 h before being subjected to confocal imaging and flow cytometry analysis. It was speculated that the uptake of these crystals might involve an active transport mechanism, which can be inhibited by low temperature, since Pos3Aa protein crystals are too large to diffuse through the cell membrane. As expected, 4° C. incubation completely abolished the uptake of Pos3Aa crystals whereas significant uptake could be observed under 37° C. (FIG. 5C, D). Given that extracellular particles can be internalized via different endocytic pathways, such as clathrin/caveolae-mediated endocytosis and macropinocytosis, a collection of known endocytic inhibitors was therefore applied to identify which pathways play dominant roles in the uptake of Pos3A crystals. A549 cells were pretreated with each inhibitor for 30 min prior to 6 h incubation with 500 nM Alexa488-Pos3Aa crystals. Uptake of Pos3Aa crystals was determined based on the percentage of Alexa488-Pos3Aa-positive cells obtained from flow cytometry analysis. Filipin III and nystatin, two inhibitors of caveolae-dependent endocytosis, resulted in no recognizable impact on the uptake of Pos3Aa crystals (FIG. 5E). Likewise, no obvious inhibition of Pos3Aa uptake was observed on the cells pretreated with chlorpromazine and dynasore, inhibitors of clathrin-mediated endocytosis (FIG. 5E). In contrast, all three inhibitors of macropinocytosis (amiloride, cytochalasin D, and nocodazole) blocked the uptake of Pos3Aa crystals by 57%, 61% and 42%, respectively (FIG. 5E). Taken together, these findings indicated that the efficient uptake of Pos3Aa protein crystals by non-phagocytic cells is mediated primarily via the macropinocytosis pathway.


3. Endosomal Escape of Pos3Aa Protein Crystals

Genetic fusion of cargo proteins to cell penetrating peptides is one of the most commonly used method to mediate the intracellular protein delivery. Despite the typically efficient endocytic uptake of CPPs, it appears that CPP-tagged proteins frequently fail to exhibit significant cellular activities. Arguably the major obstacle is the inefficient endosomal escape, which is also a challenge to micro/nano-carriers, leading to the entrapment of delivered proteins in the intracellular vesicles. To assess the ability of Pos3Aa protein crystals to escape from endo/lysosomes, A549 cells, PPFs and PC12 cells were incubated with Alexa488-Pos3Aa crystals for 24 hours and subsequently stained with LysoTracker Red DND-99 to visualize the endocytic vesicles. The co-localization analysis indicated that a large percentage of Pos3Aa crystals were localized in the cytoplasm, suggesting the occurrence of endosomal escape (FIG. 6A). To compare Pos3Aa crystals with CPPs, a cell-penetrating fusion peptide TATHA2 was fused to the C-terminus of wildtype Cry3Aa protein. Unlike Cry3Aa crystals, the resultant Cry3AaTATHA2 protein crystals can be taken up by A549 cells with the help of TATHA2 peptide (FIG. 6B). However, most of the Cry3AaTATHA2 crystals were co-localized with intracellular vesicles (FIG. 6B), indicating the incapability of TATHA2 peptide to mediate the endosomal escape. This observation is consistent with the studies on gold nanoparticles, which showed that TATHA2 peptide can only facilitate the cellular uptake but not the cytosolic localization of these particles. The near-complete colocalization of Cry3AaTATHA2 crystals with endo/lysosomes is further supported by the correlated intensity profiles of labeled crystals and LysoTrack staining, whereas the profile of Alexa488-Pos3Aa crystals displayed a distinct pattern from endo/lysosomes (FIG. 6C, D). These data provide the first evidence that Pos3Aa protein crystals have the potential as a platform for mediating the efficient cytosolic delivery of proteins.


4. Intracellular Delivery of Fluorescent mCherry Protein


To evaluate the possibility of applying Pos3Aa crystal as a carrier for protein delivery, the ability of Pos3Aa to deliver a model cargo protein, mCherry, into mammalian cells was explored. mCherry protein was genetically fused to the C-terminus of Pos3Aa, and the corresponding Pos3Aa-mCherry fusion protein crystals were produced in Bt cells and purified for subsequent experiments. Two CPP-tagged mCherry proteins, TAT-mCherry and polyarginine(R9)-mCherry, were also produced for comparison. As shown in the confocal images, Pos3Aa-mCherry crystals revealed highly efficient cellular uptake by A549 cells (FIG. 7A). The delivery efficiency was then quantified using flow cytometry. Cells treated with Pos3Aa-mCherry crystals exhibited 40-fold change in the mean mCherry fluorescence intensity, whereas the treatment of TAT-mCherry and R9-mCherry proteins only caused 1.25-fold and 1.7-fold changes, respectively (FIG. 7B, C). More importantly, significant mCherry signal was observed in the cytoplasm of cells treated with only 100 nM Pos3Aa-mCherry crystals, indicating the extremely high cellular uptake and endosomal escape efficiency of these crystals (FIG. 7D-G). Both TAT-mCherry and R9-mCherry proteins, however, were entrapped inside the endo/lysosomes, even when used in 10-fold higher concentration (1 μM) than Pos3Aa-mCherry crystals. All these results further support the suitability of Pos3Aa protein crystal as an effective platform to mediate intracellular protein delivery.


5. Intracellular Delivery of Transcription Factor p53 Protein

Given the promising results of mCherry delivery, delivery of bioactive proteins that hold potentials to be valuable therapeutics was then tested. Transcription factors are a class of key regulatory proteins controlling the eukaryotic gene expression in many biological processes, such as development and tumorigenesis. The dysfunction of transcription factors is a driver of numerous diseases, and these proteins are therefore considered to be attractive therapeutic targets. One typical example is p53 protein, whose encoding gene TP53 is found mutated or deleted in nearly half of human cancers. p53 is activated upon cellular stress signals, like oncogenic stress and DNA damage, resulting in the expression of downstream genes involved in cell-cycle arrest, DNA repair and apoptosis. Restoring p53 functions in cancer cells could be a potent alternative to cancer therapy, while the poor stability and low cell penetration efficiency of native p53 protein limit its direct intracellular delivery.


Herein, the successful delivery of p53 protein using the Pos3Aa platform is reported. Pos3Aa-p53 fusion protein was expressed in Bt Spo 407OA cells, and the resultant Pos3Aa-p53 fusion protein crystals were purified by sucrose gradient centrifugation. Live cell imaging showed that Pos3Aa-p53 crystals can be readily internalized by the p53-deficient breast cancer MDA-MB-231 cells (FIG. 8). To verify the improved uptake efficiency of p53-contained crystals in comparation with free p53 protein, a mCherry tag was utilized for visualization. As shown in FIG. 9A, obvious uptake of Pos3Aa-mCherry-p53 (Pos3Aa-mCh-p53) fusion protein crystals can be observed whereas free mCherry-p53 (mCh-p53) protein was rarely found inside MDA-MB-231 cells. Flow cytometric analysis also validated that Pos3Aa-mCh-p53 crystals were efficiently taken up by MDA-MB-231 cells in a dose-dependent manner (FIG. 9B). Given that the p53-mediated gene transcription occurs in nucleus, western blot analysis was performed to examine the nuclear localization of delivered Pos3Aa-p53 protein. As expected, Pos3Aa-p53 protein, as well as the cleaved fragment, was detected in the nuclear fraction of treated MDA-MD-231 cells (FIG. 9C, D).


The anticancer activity of Pos3Aa-p53 protein crystals was then tested using MTS assay. As indicated in FIG. 9E, Pos3Aa-p53 crystals revealed a dose-dependent inhibition of cell growth on MDA-MB-231 cells, whereas Pos3Aa-mCherry crystals or free p53 protein had no effect on the cell viability (FIG. 9E). Membrane blebbing, a defined feature of apoptosis, was observed from the Pos3Aa-p53-treated cells, supporting that p53 delivery by Pos3Aa-p53 crystals induced the apoptosis of MDA-MB-231 cells. Moreover, caspase-3/7 activity was examined using Caspase-3/7 Green Reagent, which can generate a green fluorescence upon the activation of caspase-3/7, to confirm the apoptotic activity of Pos3Aa-p53 crystals. MDA-MB-231 cells treated with 500 nM Pos3Aa-p53 crystals showed significantly higher percentage of caspase-3/7 activated cells and relative caspase-3/7 activity than cells treated with free p53 protein or Pos3Aa-mCherry crystals (FIG. 9F-H). Given that cell cycle arrest is another noticeable outcome of p53 activation, cell cycle analysis was carried out to further validate the successful delivery of p53 protein. As indicated in FIG. 9I and J, a meaningful G1 phase arrest of MDA-MB-231 cells was observed after 48 h treatment of 500 nM Pos3Aa-p53 crystals, whereas free p53 protein and Pos3Aa-mCherry crystals had no effect on the cell cycle. All these data suggest that bioactive p53 protein was effectively delivered into mammalian cells in the form of Pos3Aa-p53 fusion protein crystal.


One consequence of the p53 deficiency in cancer cells is their resistance to anticancer drugs that induce p53-dependent apoptosis. It is thus hypothesized that the restoration of p53 function by Pos3Aa-p53 crystals might be able the increase the cellular sensitivity to these anticancer drugs. To confirm this, two classic drugs, fluorouracil (5-FU) and doxorubicin (Dox) were chosen as model compounds for the subsequent validation. It has been demonstrated that loss of p53 function significantly reduces the susceptibility of cancer cells to 5-FU, whereas a p53-independent mechanism is involved in the Dox-induced cell death. It was expected that Pos3Aa-p53 treatment would make the p53-deficienct MDA-MB-231 cells more sensitive to 5-FU but not Dox. A pre-treatment experiment was first performed. Cells were pre-delivered with 500 nM Pos3Aa-p53 crystals for 24 h. After that, cells with or without pretreatment were seeded into 96-well plates at same seeding concentration, and treated with graded doses of 5-FU or Dox. As shown in FIGS. 10A and 10B, pre-treated cells (with wildtype p53) exhibited better response to 5-FU than normal MDA-MD-231 cells (with mutant p53). However, no major difference in the sensitivity to Dox was observed—supporting the success of Pos3Aa-p53-mediated delivery of wildtype p53 protein. Furthermore, combined treatment of the cells with 5-FU and Pos3Aa-p53 crystals inhibited approximately 80% of the cell growth at the highest concentration tested, while only 50% inhibition was observed for 5-FU alone (FIG. 10C). The combination index (CI) was calculated using the CompuSyn software and revealed that the CI values for 5-FU and Pos3Aa-p53 crystals at ED50, ED75, ED90 and ED95 are 0.44, 0.53, 0.66, and 0.79, indicating synergistic inhibition of cacern cell growth.


6. Intracellular Delivery of Tumor Suppressor p16 Protein

Given that the release of cargo proteins from the Cry-cargo fusion protein crystals is based on the solubilization of these crystals, a triple mutant of Pos3Aa (T533A, G535A, D536A) protein (Pos3Aa™) was generated to improve the solubility of Pos3Aa-cargo protein crystals. When compared with Pos3Aa-mCherry crystals, Pos3Aa™-mCherry protein crystals exhibited better solubility (more crystals can be solubilized) (FIG. 11). Confocal microscopy imaging showed that Pos3Aa™-mCherry crystals remain the ability to enter non-phagocytic mammalian cells (FIG. 12).


This Pos3Aa triple mutant protein was then applied to deliver another tumor suppressor protein—p16. Frequent deletions or mutations of the INK4 gene, which encodes the cyclin-dependent kinase inhibitor p16 protein, have been reported in around half of human cancers. p16 protein contributes to the regulation of cell cycle progression by binding to CDK4/6, inhibiting cyclin D-CDK4/6 complex formation and CDK4/6-mediated phosphorylation of Rb family members. The resultant hypophosphorylated Rb family members binds to E2Fs, a family of transcription factors controlling the proliferation-associated genes, prevents them from nuclear import, and consequently leads to G1 cell cycle arrest. It was hypothesized that direct delivery of p16 protein into p16-deficienct cancer cells might arrest the cells at G1 phase and thus inhibit the cell growth. To evaluate this possibility, Pos3Aa™-mCherry-p16 (Pos3Aa™-mCh-p16) fusion protein crystals were produced in Bt cells and purified using sucrose gradient centrifugation. A mCherry-tagged p16 protein (mCh-p16) was produced at the same time for comparison. p16-Deficient squamous carcinoma UM-SCC-22A cells were incubated with Pos3Aa™-mCh-p16 crystals for 24, 48 and 96 h to assess the cellular uptake of those crystals. As indicated in FIG. 13A, obvious mCherry fluorescence can still be observed after 96 h incubation. Moreover, diffused mCherry signal was clearly displayed in both cytoplasm and nucleus after 24 h (FIG. 13A), suggesting that mCherry-p16 part was successfully escaped from intracellular vesicles and released from the Pos3Aa™-mCh-p16 crystals. Flow cytometric analyses showed a dose-dependent uptake of these crystals and an much higher efficiency than free mCh-p16 protein (FIG. 13B, C). Cell cycle arrest assay was then performed to investigate if the delivered p16 protein is functional inside UM-SCC-22A cells. While 500 nM free mCh-p16 protein or Pos3Aa™-mCherry crystals had no effect on the cell cycle compared to no treatment control, more cells were arrested in G1 phase when delivered with 500 nM Pos3Aa™-mCh-p16 crystals (FIG. 13D, E). To confirm the G1 cell cycle arrest is indeed due to the restoration of p16 protein, anti-phosphorylated Rb antibodies were used to determine the level of phosphorylated Rb in UM-SCC-22A cells. Western blot results indicated that he treatment of Pos3Aa™-mCh-p16 crystals dramatically reduced the phosphorylated Rb level in UM-SCC-22A cells (FIG. 13F), demonstrating the intracellular bioactivity of delivered p16 proteins. Taken together, these findings suggest that efficient and effective delivery of p16 protein into cancer cells can be achieved by the Pos3Aa™ platform.


7. Intracellular Delivery of mCherry Protein by a Pos3Aa-Derived Peptide


It is shown that Pos3Aa crystal platform has its own advantages in intracellular delivery of functional proteins and their sustained release. Some applications, such as delivery of Cas9 protein for gene editing, however, require transient and fast action of these bioactive proteins. Hence, inspired by the rapid action of cell-penetrating peptides, a peptide derived from the domain II of Pos3Aa protein was identified and named as P3AP (FIG. 14). To test whether it can still mediate the intracellular delivery of proteins, a cargo fluorescent mCherry protein was genetically fused to the C-terminus of P3AP peptide. The soluble P3AP-mCherry protein was successfully expressed and purified from E. coli (FIG. 15). To compare its uptake efficiency with cell-penetrating peptides, the MDA-MB-231 cells were treated with 5 μM mCherry, R9-mCherry or P3AP-mCherry protein, and incubated at 37° C. for 24 h. At the end of incubation period, cells with different treatments were washed and imaged under same laser power and gain using confocal fluorescent microscopy. As shown in FIG. 16, significant internalization of mCherry by MDA-MB-231 cells was mediated by P3AP, and the efficiency was better than cell-penetrating R9 peptide. Consistently, flow cytometric analysis indicated that P3AP-mCherry exhibited the highest uptake in comparation with R9-mCherry and TAT-mCherry (FIG. 17A, B).


The abilities of cell-penetrating peptides to translocate through cell membranes is generally accompanied by cytotoxicity. To assess the cytotoxicity of P3AP peptide, MDA-MB-231 cells were incubated with different concentrations of mCherry, TAT-mCherry, R9-mCherry, and P3AP-mCherry for 72 h, and the cell viabilities were determined by MTS/PMS reagent. As indicated in FIG. 17C, P3AP-mCherry had no effect on the cell viability whereas the cell growth was significantly inhibited by R9-mCherry in a dose-dependent manner. Taken together, these results have validated that the P3AP peptide can mediate a much more efficient cellular delivery of proteins than cell-penetrating peptide with no cytotoxicity on targeted cells.


SUMMARY

Proteins are potential candidates/targets for drug research and development. While almost all current approved protein-based drugs in the market act on extracellular targets, many diseases are caused by the dysfunction of intracellular proteins. The instability and low cell penetration efficiency of proteins, however, limit the development of protein-based therapeutics. Moreover, entrapment by endo/lysosomes dramatically hinders the efficacy of protein therapeutics. To overcome these issues, the present inventors have developed a protein delivery platform based on an engineered Cry3Aa protein (Pos3Aa) with noted advantages: first, efficient endosomal escape. It has been demonstrated that a large percentage of Pos3Aa protein crystals were localized in the cytoplasm after the cellular uptake by A549 cells, primary fibroblasts (PPFs) and PC12 cells, indicating the occurrence of endosomal escape. Second, much more efficient cellular uptake than conventional cell-penetrating peptides. It has been demonstrated that cells treated with Pos3Aa-mCherry crystals exhibited 40-fold increase in the mean mCherry fluorescence intensity, whereas the treatment of TAT-mCherry and R9-mCherry proteins only caused 1.25-fold and 1.7-fold changes, respectively. Third, capability of delivering bio-functional proteins. It is further demonstrated that two tumor suppressor proteins, p53 and p16, can be efficiently delivered into cancer cells by this Pos3Aa platform. Significantly, Pos3Aa-mediated p53 and p16 delivery restored their anti-cancer activities in p53/p16-deficient cancer cells. Lastly, intracelluar delivery of proteins by a new Pos3Aa-derived peptide—P3AP. Latest results indicate that, in addition to Pos3A crystals, a P3AP peptide derived from the domain II of Pos3Aa protein can be used to efficiently deliver proteins into cells with minimal cytotoxicity.


All patents, patent applications, and other publications, including GenBank Accession Numbers, cited in this application are incorporated by reference in the entirety for all purposes.










SEQUENCE LISTING



amino acid sequence of wild-type Cry3Aa protein


SEQ ID NO: 1



MNPNNRSEHDTIKTTENNEVPTNHVQYPLAETPNPTLEDLNYKEFLRMTADNNTEALDSSTTKDVIQKGISVVGD






LLGVVGFPFGGALVSFYTNFLNTIWPSEDPWKAFMEQVEALMDQKIADYAKNKALAELQGLQNNVEDYVSALSSW





QKNPVSSPNPHSQGRIPELFSQAESHFRNSMPSFAISGYEVLFLTTYAQAANTHLFLLKDAQIYGEEWGYEKEDI





AESYKRQLKLTQEYTDHCVKWYNVGLDKLRGSSYESWVNFNRYRREMTLTVLDLIALFPLYDVPLYPKEVKTELT





RDVLTDPIVGVNNLPGYGTTFSNLENYERKPHLFDYLHPIQFHTRFQPGYYGNDSSNYWSGNYVSTRPSIGSNDI





ITSPFYGNKSSEPVQNLEFNGEKVYRAVANTNLAVWPSAVYSGVTKVEFSQYNDQTDEASTQTYDSKRNVGAVSW





DSIDQLPPETTDEPLEKGYSHQLNYVMCFLMQGSRGTIPVLTWTHKSVDFFNMIDSKKITQLPLVKAYKLQSGAS





VVAGPRFTGGDIIQCTENGSAATIYVTPDVSYSQKYRARIHYASTSQITFTLSLDGAPFNQYYFDKTINKGDTLT





YNSFNLASFSTPFELSGNNLQIGVTGLSAGDKVYIDKIEFIPVN





amino acid sequence of Pos3Aa protein


(underlined portion is P3AP sequence)


SEQ ID NO: 2



MNPNNRSEHDTIKTTENNEVPTNHVQYPLAETPNPTLEDLNYKEFLRMTADNNTEALDSSTTKDVIQKGISVVGD






LLGVVGFPFGGALVSFYTNFLNTIWPSEDPWKAFMEQVEALMDQKIADYAKNKALAELQGLQNNVEDYVSALSSW





QKNPVSSPNPHSQGRIPELFSQAESHFRNSMPSFAISGYEVLFLTTYAQAANTHLFLLKDAQIYGEEWGYEKEDI





AESYKRQLKLTQEYTDHCVKWYNVGLDKLRGSSYESWVNFNRYRREMTLTVLDLIALFPLYDVPLYPKEVKTELT





RDVLTDPIVGVNNLPGYGTTFSNLENYERKPHLFDYLHPIQFHTRFQPGYYGNDSSNYWSGNYVSTRPSIGSNDI





ITSPFYGNKSSEPVQLEFKGEKVYRAVANANTNLAVWPSAVYSGVTKVKFSQYNDTKKASKQTYDSKRNVGAVSW






DSIDQLPPETKKKPLKKGYSHQLNYVMCFLMQGSRGTIPVLTWTHKSVDFFNMIDSKKITQLPLVKAYKLQSGAS






VVAGPRFTGGDIIQCTENGSAATIYVTPDVSYSQKYRARIHYASTSQITFTLSLDGAPFNQYYFDKTINKGDTLT





YNSFNLASFSTPFELSGNNLQIGVTGLSAGDKVYIDKIEFIPVN





amino acid sequence of Pos3AaTM protein





SEQ ID NO: 3



MNPNNRSEHDTIKTTENNEVPTNHVQYPLAETPNPTLEDLNYKEFLRMTADNNTEALDSSTTKDVIQKGISVVGD






LLGVVGFPFGGALVSFYTNFLNTIWPSEDPWKAFMEQVEALMDQKIADYAKNKALAELQGLQNNVEDYVSALSSW





QKNPVSSPNPHSQGRIPELFSQAESHFRNSMPSFAISGYEVLFLTTYAQAANTHLFLLKDAQIYGEEWGYEKEDI





AESYKRQLKLTQEYTDHCVKWYNVGLDKLRGSSYESWVNFNRYRREMTLTVLDLIALFPLYDVPLYPKEVKTELT





RDVLTDPIVGVNNLPGYGTTFSNLENYERKPHLFDYLHPIQFHTRFQPGYYGNDSSNYWSGNYVSTRPSIGSNDI





ITSPFYGNKSSEPVQLEFKGEKVYRAVANANTNLAVWPSAVYSGVTKVKFSQYNDTKKASKQTYDSKRNVGAVSW





DSIDQLPPETKKKPLKKGYSHQLNYVMCFLMQGSRGTIPVLTWTHKSVDFFNMIDSKKITQLPLVKAYKLQSGAS





VVAGPRFTGGDIIQCTENGSAATIYVTPDVSYSQKYRARIHYASTSQITFTLSLDGAPFNQYYFDKTINKGDTLT





YNSFNLASFSTPFELSGNNLQIGVTGLSAGDKVYIDKIEFIPVN





amino acid sequence of CrylAa


SEQ ID NO: 4



   1 MDNNPNINEC IPYNCLSNPE VEVLGGERIE TGYTPIDISL SLTQFLLSEF VPGAGFVLGL






  61 VDIIWGIFGP SQWLAFPVQI EQLINQPIEE FARNQAISRL EGLSNLYQIY AESFREWEAD





 121 PTNPALREEM RIQFNDMNSA LTTAIPLLAV QNYQVPLLSV YVQAANLHLS VLRDVSVFGQ





 181 PWGFDAATIN SRYNDLTRLI GNYTDYAVRW YNTGLERVWG PDSRDWVRYN QFRPELTLTV





 241 LDIVALFSNY DSRRYPIRTV SQLTREIYTN PVLENFDGSF RGMAQRIEQN IPQPHLMDIL





 301 NSITIYTDVH RGFNYESGHQ ITASPVGFSG PEFAFPLFGN AGNAAPPVLV SLTGLGIFPT





 361 LSSPLYRRII LGSGPNNQEL FVLDGTEFSF ASLTTNLPST IYRQRGTVDS LDVIPPQDNS





 421 VPPRAGFSHR LSHVTMLSQA AGAVYTLRAP TFSWQHRSAE FNNIIPSSQI TQIPLTKGTN





 481 LGSGTSVVKG PGFTGGDILR RTSPGQISTL RVNITAPLSQ RYRVRIPYAS TTNLQFNTSI





 541 DGRPINQGNF SATMSSGSNL QSGSFRTVGF TTPFNFSNGS SVFTLSAHVF NSGNEVYIDR





 601 IEFVPAEVTF EAEYDLERAQ KAVNELFTSS NQIGLKTDVT DYHIDQVSNL VECLSDEFCL





 661 DEKQELSEKV KHAKPLSDER NLLQDPNFRG INRQLDRGWR GSTDITIQGG DDVFKENYVT





 721 LLGTFDECYP TYLYQKIDES KLKAYTRYQL RGYIEDSQDL EIYLIRYNAK HETVNVPGTG





 781 SLWPLSAQSP IGKCGEPNRC APHLEWNPDL DCSCPDGEKC AHHSHHFSLD IDVGCTDLNE





 841 DLGVWVIFKI KTQDGHARLG NLEFLEEKPL VGEALARVKR AEKKWRDKPE KLEWETNIVY





 901 KEAKESVDAL FVNSQYDQLQ ADTNIAMIHA ADKRVHSIRE AYLPELSVIP GVNAAIFEEL





 961 EGRIFTAFSL YDARNVIKNG DFNNGLSCWN VKGHVDVEEQ NNQRSVLVLP EWEAEVSQEV





1021 RVCPGRGYIL RVTAYKEGYG EGCVTIHEIE NNTDELKFSN CVEEEIYPNN TVTCNDYTVN





1081 QEEYGGAYTS RNRGYNEAPS VPSFYSDVYE EKSYTDGRRE NPCEFNRGYR DYTPLPVGYV





1141 TKELEYFPET DKVWIEIGET EGTFIVDGVE LLLMEE





amino acid sequence of Cry2Aa


SEQ ID NO: 5



   1 MNNVLNGGRT TICDAYNVVA HDPFSFEHKS LDTIQKEWME WKPTDHSLYV APVVGTVSGF






  61 LLKKVGSLIG KRILSELWGI IFPSGSTNLM QDILRETEQF LNQRLNTDTL ARVNAELIGL





 121 QANIREFNQQ VDNFLNPTQN PVPLSITSSV NTMQQLFLNP LPQFQIQGYQ LLLLPLFAQA





 181 ANMHLSFIRD VILNSFEWGI SAATLRTYRD YLRNYTRDYS NYCINTYQYQ FRGLNTRLHD





 241 MLEFRTYMFL NVFEYVSIWS LFKYQSLMVS SGANLYAGGS GPQQTQSFTA QNWPFLYSLF





 301 QVNSNYILSG ISGTRLSITF PNIGGLPGGT TTHSLNSARV NYSGGVSSGL IGATNLNHNP





 361 NCSTVLPPLS TPFVRSWLDS GTDREGVATS TNWQTESFQT TLSLRCGAFS ARGNSNYFPD





 421 YFIPNISGVP LVIRNEDLTP PLHYNQIRNI ESPSGTPGGA PAYLVSVHNR KNNIYAANEN





 481 GTMIHLAPED YTGFTISPIH ATQVNNQTRT FIGEKFGNQG DSLRFEQSNT TARTTLRGNG





 541 NSYNLYLPVS SIGNSTIRVT INGRVYTVSN VNTTTNNDGV NDNGARFSDI NIGINVASDN





 601 TNVTLDINVT LNSGTPFDLM NIMFVPTNLP PLY





amino acid sequence of Cry4Aa


SEQ ID NO: 6



   1 MNPYQNKNEY ETLNASQKKL NISNNYTRYP IENSPKQLLQ STNYKDWLNM CQQNQQYGGD






  61 FETFIDSGEL SAYTIVVGTV LTGFGFTTPL GLALIGFGTL IPVLFPAQDQ SNTWSDFITQ





 121 TKNIIKKEIA STYISNANKI LMRSFNVIST YHNHLKTWEN NPNPQNTQDV RTQIQLVHYH





 181 FQNVIPELVN SCPPNPSDCD YYNILVLSSY AQAANLHLTV LNQAVKFEAY LKNNRQFDYL





 241 EPLPTAIDYY PVLTKAIEDY TRYCVTTYKK GLNLIKTTPD SNLDGNINWN TYNTYRTKMT





 301 TAVLDAVVLF PNYDVGKYPI GVQSELTREI YQVLNFEESP YKYYDFQYQE DSLTRRPHLF





 361 TWLDSLNFYE KAQTTPNNFF TSHYNMFHYT LDNISQKSSV FGNHNVTDKL KSLGLATNIY





 421 IFLLNVISLD NKYLNDYNNI GKMDFFITNG TRLLEKELTA GSGQITYDVN KNIFGLPILK





 481 RRENQGNPTL FPTYDNYSHI LSFIKSLSIP ATYKTQVYTF AWTHSSVDPK NTIYTH1TTQ





 541 IPAVKANSLG TASKVVQGPG HTGGDLIDFK DHFKITCQHS NFQQSYFIRI RYASNGSANT





 601 PAVINLSIPG VAELGMALNP TFSGTDYTNL KYKDFQYLEF SNEVKFAPNQ NISLVFNRSD





 661 VYTNTTVLID KIEFLPITPS IREDREKQKL ETVQQIINTF YANPIKHTLQ SELTDYDIDQ





 721 AANLVECISE ELYPKEKMLL LDEVKNAKQL SQSRNVLQKG DFESATLGWT TSDNITIQED





 781 DPIFKGHYLH MSGARDILGT IFPTYIFQKI DESKLKPYTR YLVPGFVGSS KDVELVVSRY





 841 GEEIDAIMNV FADLNYLYPS TFDCEGSNRC ETSAVPANIG NTSDMLYSCQ YDTGKKHVVC





 901 QDSHQFSFTI DTGALDTNEN IGVWVMFKIS SPDGYASLDN LEVIEEGPID GEALSPVKHM





 961 EKKWNDQMEA KRSETQQAYD VAKQAIDALF TNVQDEALQF DTTLAQIQYA EYLVQSIPYV





1021 YNDWLSDVPG MRYDIYVELD APVAQARYLY DIRNIIKNGD FTQGVMGWHV TGNADVQQID





1081 GVSVLVLSNW SAGVSQNVHL QHNHGYVLGV IAKKEGPGNG YVTLMDWEEN QEKLTFTSCE





1141 EGYITKTVDV FPDTDRVPIE IGETEGSFYI ESIELICMNE





amino acid sequence of Cry5B


SEQ ID NO: 7



   1 MATINELYPV PYNVLAHPIK EVDDPYSWSN LLKGIQEGWE EWGKTGQKKL FEDHLTIAWN






  61 LYKTGKLDYE ALTKASTSLI GFTPGAEAAV PFINMFVDFV WPKLFGANTE GKDQQLFNAI





 121 MDAVNKMVDN KFLSYNLSTL NKTIEGLQGN LGLFQNAIQV AICQGSTPER VNFDQNCTPC





 181 NPNQPCKDDL DRVASRFDTA NSQFTQHLPE FKNPWSDENS TQEFKPTSVE LTLPMYTTVA





 241 TLHLLLYEGY IEFMTKWNFH NEQYLNNLKV ELQQLIHSYS ETVRTSFLQF LPTLNNPSKS





 301 SVNAYNRYVR NMTVNCLDIA ATWPTFDTHN YHQGGKLDLT RIILSDTAGP IEEYTTGDKT





 361 SGPEHSNITP NNILDTPSPT YQHSFVSVDS IVYSRKELQQ LDIATYSTNN SNNCHPYGLP





 421 LSYTDGSRYD YGDNQPDFTT SNNNYCHNSY TAPITLVNAR HLYNAKGSLQ NVESLVVSTV





 481 NGGSGSCICD AWINYLRPPQ TSKNESPPDQ KINVLYPITE TVNKGTGGNL GVISAYVPME





 541 LVPENVIGDV NADTKLPLTQ LKGFPFEKYG SEYNNRGISL VPEWINGNNA VKLSNSQSVG





 601 IQITNQTKQK YEIRCRYASK GDNNVYFNVD LSENPFPNSI SFGSTESSVV GVQGENGRYI





 661 LKSITTVEIP AGSFYVHITN QGSSDLFLDR IEPVPKIQFQ FCDNNNLHCD CNNPVDTDCT





 721 FCCVCTSLTD CDCNNPRGLD CTLCCQVENQ LPSFVTLTDL QNITTQVNAL VASSEHDTLA





 781 TDVSDYEIEE VVLKVDALSG EVFGKEKKAL RKLVNHTKRL SKARNLLIGG NFDNLDAWYR





 841 GRNVVNVSDH ELFKSDHVLL PPPTLYSSYM FQKVEESKLK ANTRYTVSGF IAHAEDLEIV





 901 VSRYGQEVKK VVQVPYGEAF PLTSPGAICC PPRSTSNGKP ADPHFFSYSI DVGTLLVEAN





 961 PGIELGLRIV ERTGMARVSN LEIREDPPLK KNELRNVQRA ARNWRTATDQ ERAEVTALIQ





1021 PVLNQINALY ENEDWNGAIP SGVSYHDLEA IVLPTLPKLN HWFMSDMLGE QGSILAQFQE





1081 ALDRATTQLE ESTILHNGHF TTDAANWTIE GDAHHAILED GRRVLRLPDW SSSVSQTIEI





1141 ENFDPDKEYQ LVFHAQGEGT VSLQHGEEGE YVETHPRKSA NFTTSHRQGV TFETNKVTVE





1201 ITSEDGEFLV DHIALVEAPL PTDDQSSDGN TTSNTNSNTS MMNNQ





amino acid sequence of Cry7Ca1


SEQ ID NO: 8



   1 MDKQNDSGII KATLNEDFSN SIQRYPLVTD QTINYKDFLN MNEEIAPYAS SKDVIFSSIS






  61 IIRTEMGFAG HGTAGGIIGL FTEVLRLLWP NKQMDLWESF MNEVEALINQ EITEAVVSKA





 121 LSELEGLPNA LEGYTSALEA WQNNRSDKLK QLLVYERFVS TENLFKFAMP SFRSVGFEGF





 181 LLTVYAQAAN LHLFLLKNAE LFGAEWGMQQ YEIDLFYNEQ KGYVEEYTDR CVKWYKEGLN





 241 KLKNASGVKG KVWENYNRFP REMTIMVLDL LPLFPIYDAR TYPMETVTEL TRQIFTDPIG





 301 LTGINETKYP DWYGAASSEF VLIENPAIPK PGLFQWLTKI NVRARVVEPN DRFAIWTRHS





 361 VVTQCTKSTT ENTFNYGTSS GSTLSHTFDL LSKDIYQTYS IAAANKSATW YQAVPLLRLY





 421 GINSSNVLSE DAFSFSNNIP SSKCKSTYSS DQLPIELLDE PIYGDLEEYG RRLSYVSEIF





 481 KETGSGTIPV LGWTHVSVRP DNKLYPDKIT QIPAVKAFET NTAGVEIIDS ASTGGPILKI





 541 VNNNLPSNQV FPMPLSFSEP QKTKVRVRYA ATGDGVMSFS GIAHDEYFTA TMKEGEALKY





 601 SYLTMGNDYA GTAAELSMIY TTKANTSNCT IYIDKIEEIP VDENYNNRVQ LEKAQRAVNT





 661 LFTAGRNALQ KDVTDFKVDQ VSILVDCVSG ELYPNEKREL LSLVKYAKPL SYSRNLLLLP





 721 TFDSINSSEE NGWNGSNGIA IGSGDFVFKG NYLIFSGTND EQYPTYLYQK IDESKLKEYT





 781 PYKLRGFIES SQDLEAYVIR YDAKHETLDV SMNLLPDIPP VNACGEPNRC AALQYLDENP





 841 KLECSSIQDG ILSDSHSFSL HIDTGSIDFN ENVGIWVLFK ISTPEGYAKF GNLEVIEDGP





 901 VIGEALARVK RQETKWRNKL TQLRTETQAI YTRAKQALDN LFTDAQDSHL KIGATFAAIV





 961 AARKIVQSIR EAYMSWLSDV PGLNYPIFTE LNDRVQRAFQ LYDVQNVVRN GRFLNGVLLW





1021 IVTSDVRVQE GNGNNVLVLS GWDAQVLQCL NLYQNRGYIL RVTARKEGLG EGYITITDEE





1081 GNTDQLTFGS CENIDSSNSF VSTGYITKEL EFFPDTDQIQ IEIGETEGTF QVESVELFLM





1141 ENLC





amino acid sequence of Cry8Ea1


SEQ ID NO: 9



MSPNNQNEYEIIDMAPSTSVSNDSNRYPFASDPTNALQNMNYKEYLRMSEGYDSEYSGSP






EVLISERDAVKTAISLVGTILGKLGVPLVGPIVSLYSTLIDVLWPGGKSQWEIFMEQVEA





LINQKIAEYARAKALAELEGLGNNYQLYLTALEEWQENPSSTRVLRDVPNRFEILDSLFT





QYMPSFRVTGYEVPLLSVYAQAANLHLLLLKDASIFGEEWGFSTTAINNYYNRQMSLIAQ





YSDHCVQWYRTGLDRLKGSNEKQWVEYNRFPREMTLSVLDIMTLFPMYDMRTYPMETKAQ





LTREVYTDPIGAIGAQGSWYDSAPSFNTLESTSIRGKHLFDFITRLSIYTGRSSSSASNY





YYGVSKVVFDAIYPDNKYKTTFTYNPGGEGIGAQEKDSEVELPPETLDQPNYEATSHRLN





YVTFIRNPDVPVFSWTHPSADRTNTVYSDKITQIPVVKASDGPKPSANEVGHYLGGDPIS





FNSSGSTGVIRLNINSPLSQKYRVRIRYCSSVDFDLDVVPGGTTVNNGPFNKSAPNVGWQ





SLKYENFKFASFSTPFTFNQAQDTLKISVRNFSSIVGGSVVYIDRIELIPVNATYEAEQD





LDSAKKAVNTLFTNTKDGLRPGVTDYEVNQAANLVECLSDDLYPNEKRLLFDAVKEAKRL





SEAPNLLQDPDFQEINGENGWTASTGIEVVEGDALFKGRYLRLPGAREMDTETYPTYLYQ





KVEEGVLKPYTRYRLRGFVGSSQGLEISTIRHQTNRIVKNVPDDLLPDVPPVNSDGRINR





CSEQKYVNSRLEGERGLPNGNRSAEAHEFSLPIDIGELDYNENAGIWVGFKITDPEGYAT





LGNLELVEEGPLSGDALERLQREEQQWKLQMTKRREETDRKYTAAKQAVDRKYADYDDQQ





LNPNVEITDITAAQNLIQSIPYVYNEMFPEIQGMNYTKYTELTNRLQQAWGLYDQPNAIP





NGDFPNELSNWNTTSGVNVQQINNTSVLVMPNWDGQVSQQFTVQPNQRYVLRVTARKEGV





GNGYVSIRDGGNQTETLTFSASDYNTDSVYNTQVSNTNGLYNEQTGYTTKTVTFIPYTDQ





VWIEMSETGFMFYIESVELIVDVE





amino acid sequence of Cry10Aa


SEQ ID NO: 10



MYPYQNKNEYEIFNAPSNGFSKSNNYSPYPLANKPNQPLKNTNYKDWLNVCQDNQQYGNN






AGNFASSETIVGVSAGIIVVGTMLGAFAAPVLAAGIISFGTLLPIFWQGSDPANVWQDLL





NIGGRPIQEIDKNIINVLTSIVTPIKNQLDKYQEFFDKWEPARTHANAKAVHDLFTTLEP





IIDKDLDMLKNNASYRIPTLPAYAQIATWHLNLLKHAATYYNIWLQNQGINPSTFNSSNY





YQGYLKRKIQEYTDYCIQTYNAGLTMIRTNTNATWNMYNTYRLEMTLTVLDLIAIFPNYD





PEKYPIGVKSELIREVYTNVNSDTFRTITELENGLTRNPTLFTWINQGRFYTPNSRDILD





PYDIFSFTGNQMAFTHTNDDPNIIWGAVHGNIISQDTSKVFPFYRNKPIDKVEIVRHREY





SDIIYEMIFFSNSSEVFRYGGNSTIENNYKRTDSYMIPKQTWKNEEYGHTLSYIKTDNYI





FSVVRERRPVAFSWTHTSVDFQNTIDLLNITQIHALKALKVSSDSKIVKGPGHTGGDLVI





LKDSMDFRVRFLKNVSRQYQVRIRYATNAPKTTVFLTGIDTISVELPSTTSRQNPNATDL





TYADFGYVTFPPTVPNKTFEGEDTLLMTLYGTPNHSYNIYIDKIEFIPITQSVLDYTEKQ





NIEKTQKIVNDLFVN





amino acid sequence of Cry11Aa


SEQ ID NO: 11



MEDSSLDTLSIVNETDFPLYNNYTEPTIAPALIAVAPIAQYLATAIGKWAAKAAFSKVLS






LIFPGEQPATMEKVRTEVETLIEQKLSQERVNILNAEYRGIIEVSDVFDAYIKQPGFTPA





VDSFIKLFNQKVLDYRTRLMRMYTEEFGRLCKVSLKDGLTFRNMCNLYVFPFAEAWSLMR





YELGLKLSQSSLWDYVGVSIPVNYVEWGGLVYKLLMGEVNQRLTTVKFNYSFTNEPADIP





ARENIRGVHPIYDPSSGLTGWIGNGRTNNFNFADNNGNEIMEVRTQTFYQNPNNEPIAPR





DIINQILTAPAPADLFFKNADINVKFTQWFQSTLYGWNIKLGTQTVLSSRTGTIPPNYLA





YDGYYIRAISACPRGVSLAYNHDLTTLTYNRIEYDSPTTENIIVGFAPDNTKDFYSKKSH





YLSETNDSYVIPALQFAEVSDRSFLEDTPDQATDGSIKFARTFISNEAKYSIRLNTGFNT





ATRYKLIIRVRVPYRLPAGIRVQSQNSGNNRMLGSFTANANPEWVDFVTDAFTFNDLGIT





TSSTNALFSISSDSLNSGEEWYLGQLFLVKEGAFTTQINPLLK





Claims
  • 1. A polypeptide comprising a modified SEQ ID NO:1, with two or more, five or more, or all of amino acids at residues 391, 395, 423, 430, 432, 433, 436, 461, 462, 463, and 466 of SEQ ID NO:1 replaced with lysine, wherein the polypeptide forms crystal upon being expressed in a host cell.
  • 2-7. (canceled)
  • 8. A nucleic acid comprising a polynucleotide sequence encoding the polypeptide of claim 1.
  • 9. An expression cassette comprising a promoter operably linked to a polynucleotide sequence encoding the polypeptide of claim 1.
  • 10. A vector comprising the expression cassette of claim 9.
  • 11. A host cell comprising the polypeptide of claim 1.
  • 12-13. (canceled)
  • 14. A method for recombinantly producing a polypeptide, comprising the steps of (i) introducing the nucleic acid of claim 8; and (ii) culturing the cell under conditions permissible for the expression of the fusion protein.
  • 15-17. (canceled)
  • 18. A composition comprising the polypeptide of claim 1 and a mammalian cell.
  • 19-20. (canceled)
  • 21. A method delivering a therapeutic protein into a mammalian cell, comprising the step of contacting the polypeptide of claim 1 with the mammalian cell, wherein the polypeptide is a fusion protein of the modified SEQ ID NO:1 and the therapeutic protein, and the polypeptide is crystalized.
  • 22-23. (canceled)
  • 24. A soluble polypeptide comprising a fragment of SEQ ID NO:2, wherein the fragment is no longer than about 150 amino acids and comprises the 394-466 segment of SEQ ID NO:2.
  • 25-27. (canceled)
  • 28. A nucleic acid comprising a polynucleotide sequence encoding the soluble polypeptide of claim 24.
  • 29. An expression cassette comprising a promoter operably linked to a polynucleotide sequence encoding the soluble polypeptide of claim 24.
  • 30. A vector comprising the expression cassette of claim 29.
  • 31. A host cell comprising the soluble polypeptide of of claim 24.
  • 32-33. (canceled)
  • 34. A method for recombinantly producing a soluble polypeptide, comprising the steps of (i) introducing the nucleic acid of claim 28; and (ii) culturing the cell under conditions permissible for the expression of the soluble polypeptide.
  • 35-37. (canceled)
  • 38. A composition comprising the soluble polypeptide of claim 24 and a mammalian cell.
  • 39. (canceled)
  • 40. A method delivering a therapeutic protein into a mammalian cell, comprising the step of contacting the soluble polypeptide of claim 24 with the mammalian cell, wherein the soluble polypeptide is a fusion protein of the fragment of SEQ ID NO:2 and the therapeutic protein.
  • 41-42. (canceled)
RELATED APPLICATIONS

This application claims priority to U.S. Patent Application No. 62/834,605, filed Apr. 16, 2019, the contents of which are hereby incorporated by reference in the entirety for all purposes.

PCT Information
Filing Document Filing Date Country Kind
PCT/CN2020/084939 4/15/2020 WO
Provisional Applications (1)
Number Date Country
62834605 Apr 2019 US